1
|
Smith HL, Broszczak DA, van der Burg CA, Surm JM, Liggins L, Norton RS, Prentis PJ. A comparative analysis of toxin gene families across diverse sea anemone species. Toxicon X 2025; 26:100217. [PMID: 40162058 PMCID: PMC11952004 DOI: 10.1016/j.toxcx.2025.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/03/2025] [Accepted: 02/23/2025] [Indexed: 04/02/2025] Open
Abstract
All species from order Actiniaria (sea anemones) are venomous, even though most are of no threat to humans. Currently, we know very little about the toxin gene complement of highly venomous members of this order. To address this gap in knowledge, we sequenced the transcriptome of the highly venomous and medically significant Hell's Fire sea anemone, Actinodendron plumosum, as well as five distantly related species, Cryptodendrum adhaesivum, Epiactis australiensis, Heteractis aurora, Isactinia olivacea and Stichodactyla mertensii. We used bioinformatic approaches to identify their toxin gene complements and performed a comparative evolutionary analysis of seven understudied toxin families. Of the 16 toxin families identified, 12-40 candidate toxins were found in the six new sea anemone transcriptomes, with only 12 candidates in eight toxin families identified in A. plumosum. Across 26 sea anemone species, six neurotoxin families showed evidence of taxonomic restriction, whereas the phospholipase A2 toxin family was ubiquitously distributed. Additionally, we identified two alternative forms for the phospholipase A2 toxin family, a 10- and 14-cysteine framework, which warrant further structural and functional characterisation. Overall, we have identified a comprehensive list of toxins from a wide diversity of sea anemone species that provides the basis for future research to structurally and functionally characterise novel candidates for potential use as therapeutics or for agricultural applications.
Collapse
Affiliation(s)
- Hayden L. Smith
- School of Biology and Environmental Sciences, Faculty of Science, Queensland University of Technology, Brisbane, Australia
| | - Daniel A. Broszczak
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4000, Australia
| | - Chloé A. van der Burg
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, 9016, New Zealand
| | - Joachim M. Surm
- Faculty of Biology, Ludwig-Maximilians-Universität Munich, Munich, D-80539, Germany
| | - Libby Liggins
- School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Raymond S. Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria, 3052, Australia
| | - Peter J. Prentis
- School of Biology and Environmental Sciences, Faculty of Science, Queensland University of Technology, Brisbane, Australia
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
2
|
Canak HN, Bas K, Yağmur EA, Karakurt S. Mesobuthus eupeus venom modulates colorectal carcinoma signaling pathways and induces apoptosis. Med Oncol 2025; 42:163. [PMID: 40229568 PMCID: PMC11996983 DOI: 10.1007/s12032-025-02689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Colorectal cancer (CRC) is a significant global health concern, often challenging to treat effectively with conventional methods and burdened by adverse effects. Scorpion venoms offer a unique avenue for exploration, given their ability to disrupt the cell cycle, inhibit growth, and trigger apoptosis. This study delves into the impact of Mesobuthus eupeus (M. eupeus) scorpion venom on the proliferation and progression of colorectal cancer at the molecular level. The total protein concentration in the venom (607.5 µg/mL) also emphasized the rich composition and potential for therapeutic applications. The study reveals that M. eupeus venom effectively reduced the proliferation of DLD-1 and HT-29 colorectal cancer cells in a dose-dependent manner with IC50 values of 4.32 and 7.61 µg/mL, respectively. The venom also impedes cell migration, diminishes colony formation, and triggers apoptosis in the cancer cells. The venom also induced early and late apoptosis in the two cancer cell lines. The human colorectal cancer and apoptotic pathways were clarified at the molecular level using pathway panels, which revealed that 16 genes involved in colorectal cancer increased while 23 decreased. In the HT-29 cell line, 57 genes increased, and 1 decreased following venom treatment. Besides, the mRNA expression of 19 genes involved in the apoptotic pathway was increased, while 22 were reduced in DLD-1 cells. This study underscores the potential of M. eupeus venom as a natural therapeutic approach in the quest for cancer treatments.
Collapse
Affiliation(s)
- Havva Nur Canak
- Faculty of Science, Department of Biochemistry, Selcuk University, Konya, Türkiye
| | - Kemal Bas
- Faculty of Science, Department of Biochemistry, Selcuk University, Konya, Türkiye
| | - Ersen Aydın Yağmur
- Department of Plant and Animal Production, Alasehir Vocational High School, Manisa Celal Bayar University, Manisa, Türkiye
| | - Serdar Karakurt
- Faculty of Science, Department of Biochemistry, Selcuk University, Konya, Türkiye.
| |
Collapse
|
3
|
Hsiao MH, Miao Y, Liu Z, Schütze K, Limjunyawong N, Chien DCC, Monteiro WD, Chu LS, Morgenlander W, Jayaraman S, Jang SE, Gray JJ, Zhu H, Dong X, Steinegger M, Larman HB. Molecular Display of the Animal Meta-Venome for Discovery of Novel Therapeutic Peptides. Mol Cell Proteomics 2025; 24:100901. [PMID: 39746545 PMCID: PMC11833617 DOI: 10.1016/j.mcpro.2024.100901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/20/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
Animal venoms, distinguished by their unique structural features and potent bioactivities, represent a vast and relatively untapped reservoir of therapeutic molecules. However, limitations associated with comprehensively constructing and expressing highly complex venom and venom-like molecule libraries have precluded their therapeutic evaluation via high-throughput screening. Here, we developed an innovative computational approach to design a highly diverse library of animal venoms and "metavenoms". We used programmable M13 hyperphage display to preserve critical disulfide-bonded structures for highly parallelized single-round biopanning with quantitation via high-throughput DNA sequencing. Our approach led to the discovery of Kunitz-type domain containing proteins that target the human itch receptor Mas-related G-protein coupled receptor member X4, which plays a crucial role in itch perception. Deep learning-based structural homology mining identified two endogenous human homologs, tissue factor pathway inhibitor (TFPI), and serine peptidase inhibitor, Kunitz type 2 (SPINT2), which exhibit agonist-dependent potentiation of Mas-related G-protein coupled receptor member X4. Highly multiplexed screening of animal venoms and metavenoms is therefore a promising approach to uncover new drug candidates.
Collapse
Affiliation(s)
- Meng-Hsuan Hsiao
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yang Miao
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zixing Liu
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Biology, Zanvyl Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Konstantin Schütze
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Center of Research Excellence in Allergy and Immunology, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Daphne Chun-Che Chien
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wayne Denis Monteiro
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lee-Shin Chu
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - William Morgenlander
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sahana Jayaraman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sung-Eun Jang
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Martin Steinegger
- School of Biological Sciences, Seoul National University, Seoul, South Korea; Artificial Intelligence Institute, Seoul National University, Seoul, South Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.
| | - H Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
4
|
Stanajic-Petrovic G, Keck M, Barbe P, Urman A, Correia E, Isnard P, Duong Van Huyen JP, Chmeis K, Diarra SS, Palea S, Theodoro F, Nguyen AL, Castelli F, Pruvost A, Zhao W, Mendre C, Mouillac B, Bienaimé F, Robin P, Kessler P, Llorens-Cortes C, Servent D, Nozach H, Maillère B, Guo D, Truillet C, Gilles N. A Snake Toxin Derivative for Treatment of Hyponatremia and Polycystic Kidney Diseases. J Am Soc Nephrol 2025; 36:181-192. [PMID: 39431458 PMCID: PMC11801765 DOI: 10.1681/asn.0000000505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Key Points MQ232, a disulfide-bond reticulated peptide derived from a natural snake toxin, was optimized as a new aquaretic drug candidate. MQ232 showed very low acute and chronic toxicity in rat and a biodistribution in mice strongly in favor of the kidney organs. MQ232 induced a sole aquaretic effect and demonstrated high in vivo activities on hyponatremia and polycystic kidney disease models. Background Vaptans were developed at the end of the previous century as vasopressin type 2 receptor antagonists. Tolvaptan is the most prescribed vaptan for hyponatremia and autosomal dominant polycystic kidney disease (ADPKD). However, its use is not as widespread as it should be due to price issues, a narrow therapeutic window, and some side effects. With the aim of discovering new efficient and safer vasopressin type 2 receptor antagonists, we screened animal venoms and identified several peptide toxins. Among them, mambaquaretin 1 (MQ1) displayed unique biological properties in that regard that it was the starting point for the development of a potential drug candidate. Methods Human T-cell assays and bioinformatics were used to mitigate MQ1 immunogenicity risk. MQ232 biodistribution in mice was performed by positron emission tomography. Pharmacodynamics, pharmacokinetics, and acute and chronic toxicity tests were performed on control rats. A rat experimental model of desmopressin-induced hyponatremia, ex vivo mice model of kidney cysts, and mice orthologous model of ADPKD were used to validate MQ232 efficacy in these pathologies. Results Three mutations were introduced in MQ1 to mitigate its immunogenicity risk. A fourth gain-of-function mutation was added to generate MQ232. MQ232's safety was demonstrated by a first toxic dose as high as 3000 nmol/kg and a strong kidney organ selectivity by positron emission tomography imaging, while showing almost no interaction with the liver. MQ232's efficacy was first demonstrated with an effective dose of 3 nmol/kg in a hyponatremic model and then in polycystic kidney models, on which MQ232 significantly reduced cyst growth. Conclusions We demonstrated, using diverse translational techniques and minimizing animal use, MQ232's safety and efficacy in several rodent models of hyponatremia and ADPKD.
Collapse
Affiliation(s)
- Goran Stanajic-Petrovic
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Mathilde Keck
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Peggy Barbe
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Apolline Urman
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Evelyne Correia
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Pierre Isnard
- Anatomie et Cytologie Pathologiques, CHU Necker-Enfants Malades, Paris, France
| | | | - Khawla Chmeis
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | | | - Stefano Palea
- Humana Biosciences, Prologue Biotech, Labège, France
| | - Frederic Theodoro
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Anvi-Laëtitia Nguyen
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Florence Castelli
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Alain Pruvost
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Wenchao Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | | | - Bernard Mouillac
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frank Bienaimé
- Service d'Explorations Fonctionnelles, Département Croissance et Signalisation, Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Robin
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Pascal Kessler
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Catherine Llorens-Cortes
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Hervé Nozach
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Bernard Maillère
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Charles Truillet
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Nicolas Gilles
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| |
Collapse
|
5
|
Cordeiro BM, Leite Fontes CF, Meyer-Fernandes JR. Molecular Basis of Na, K-ATPase Regulation of Diseases: Hormone and FXYD2 Interactions. Int J Mol Sci 2024; 25:13398. [PMID: 39769162 PMCID: PMC11678576 DOI: 10.3390/ijms252413398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The Na, K-ATPase generates an asymmetric ion gradient that supports multiple cellular functions, including the control of cellular volume, neuronal excitability, secondary ionic transport, and the movement of molecules like amino acids and glucose. The intracellular and extracellular levels of Na+ and K+ ions are the classical local regulators of the enzyme's activity. Additionally, the regulation of Na, K-ATPase is a complex process that occurs at multiple levels, encompassing its total cellular content, subcellular distribution, and intrinsic activity. In this context, the enzyme serves as a regulatory target for hormones, either through direct actions or via signaling cascades triggered by hormone receptors. Notably, FXYDs small transmembrane proteins regulators of Na, K-ATPase serve as intermediaries linking hormonal signaling to enzymatic regulation at various levels. Specifically, members of the FXYD family, particularly FXYD1 and FXYD2, are that undergo phosphorylation by kinases activated through hormone receptor signaling, which subsequently influences their modulation of Na, K-ATPase activity. This review describes the effects of FXYD2, cardiotonic steroid signaling, and hormones such as angiotensin II, dopamine, insulin, and catecholamines on the regulation of Na, K-ATPase. Furthermore, this review highlights the implications of Na, K-ATPase in diseases such as hypertension, renal hypomagnesemia, and cancer.
Collapse
Affiliation(s)
- Bárbara Martins Cordeiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Carlos Frederico Leite Fontes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| |
Collapse
|
6
|
Xian T, Cao M, Chen K, Zhao W, Liu Y, Yao W, Guang H, Yang Y, Su M, Zhang R, Ma J, Ma L, Gao J. Identification of a novel protein Hq023 of the hard tick Haemaphysalis qinghaiensis and preliminary evaluation of its analgesic effect in mice model. Parasitol Int 2024; 103:102933. [PMID: 39048024 DOI: 10.1016/j.parint.2024.102933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
Tick saliva contains a range of critical biological molecules which could inhibit host defenses and guarantee their food supply. Hq023, a novel cDNA sequence, was cloned from a cDNA library constructed from salivary glands of partially-engorged Haemaphysalis qinghaiensis. Hq023 has an open reading frame (ORF) of 408 bp coding a protein containing 135 amino acid residues with a molecular mass of 15 kDa. Database homology showed that Hq023 protein was structurally similar to a natural toxin U33-theraphotoxin-Cg1c from the Chinese tarantula Chilobrachys guangxiensis. A recombinant protein was expressed with the novel cDNA in a prokaryotic system and its analgesic effect was evaluated in mice model. Both tail immersion and hot-plate tests uncovered an antinociceptive activity, while in the acetic acid-induced writhing test this effect was not observed. These results indicated that the novel recombinant protein Hq023 (rHq023) probably possessed a central antinociceptive activity. Finding of the novel protein might pave a new avenue for the development of tick-derived analgesics.
Collapse
Affiliation(s)
- Tong Xian
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China; Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Meina Cao
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Kaiting Chen
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Wenbin Zhao
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Yueqing Liu
- Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Wenjing Yao
- Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Hui Guang
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Yinran Yang
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Muya Su
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Ruijuan Zhang
- Department of Pharmacy, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China
| | - Jing Ma
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China; Third Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450003, China
| | - Linyuan Ma
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China; Ordos Clinical Medical College, Inner Mongolia Medical University, Ordos 017000, China
| | - Jinliang Gao
- Laboratory of Molecular Medicine, Ordos Central Hospital, Inner Mongolia Autonomous Region, Ordos 017000, China; Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014000, China; Ordos Clinical Medical College, Inner Mongolia Medical University, Ordos 017000, China.
| |
Collapse
|
7
|
Jeong H, Jang S, Park JK, Kim KH, Park JH, Lee G, Sung SH. Bee Venom Acupuncture for Shoulder Pain: A Literature Review of Clinical Studies. Toxins (Basel) 2024; 16:501. [PMID: 39591256 PMCID: PMC11598799 DOI: 10.3390/toxins16110501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Managing shoulder pain typically involves the use of acetaminophen or oral nonsteroidal anti-inflammatory drugs, but prolonged use of these medications can lead to dependence and various side effects. To overcome the dose dependency and side effects of these conventional drugs, animal venoms have begun to be utilized. Among them, bee venom stands out for its powerful anti-inflammatory properties, which help relieve pain and treat chronic inflammatory conditions. This review evaluates the efficacy and safety of bee venom acupuncture (BVA) for shoulder pain. In March 2024, we searched 11 databases: 5 international and 6 Korean databases. We identified 23 clinical studies on BVA for shoulder pain. The causes of shoulder pain were post-stroke pain (43.5%), rotator cuff syndrome (17.4%), and brachial plexus palsy (13.0%). The BVA concentration and dosage per session were 0.005-1.0 mg/mL and 0.01-2.0 mL, respectively. All included clinical studies reported positive effects on pain outcomes. This review suggests that BVA, which involves injecting bee venom into acupuncture points, may serve as a viable alternative for pain management. However, the level of evidence in the included studies was low and adverse effects were reported infrequently, indicating that further research is needed.
Collapse
Affiliation(s)
- Hyein Jeong
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Soobin Jang
- Department of Preventive Medicine, College of Korean Medicine, Daegu Haany University, 1 Haanydaero, Gyeongsan 38610, Republic of Korea;
| | - Jang-Kyung Park
- Department of Korean Medicine Obstetrics and Gynecology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Kyeong Han Kim
- Department of Preventive Medicine, College of Korean Medicine, Woosuk University, Jeonju 54986, Republic of Korea;
| | - Jong Hyun Park
- Department of Pathology, College of Korean Medicine, Daegu Haany University, 1 Haanydaero, Gyeongsan 38610, Republic of Korea;
| | - Gihyun Lee
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea
| | - Soo-Hyun Sung
- Department of Policy Development, National Institute for Korean Medicine Development, Seoul 04516, Republic of Korea
| |
Collapse
|
8
|
Wang CR, McFarlane LO, Pukala TL. Exploring snake venoms beyond the primary sequence: From proteoforms to protein-protein interactions. Toxicon 2024; 247:107841. [PMID: 38950738 DOI: 10.1016/j.toxicon.2024.107841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
Snakebite envenomation has been a long-standing global issue that is difficult to treat, largely owing to the flawed nature of current immunoglobulin-based antivenom therapy and the complexity of snake venoms as sophisticated mixtures of bioactive proteins and peptides. Comprehensive characterisation of venom compositions is essential to better understanding snake venom toxicity and inform effective and rationally designed antivenoms. Additionally, a greater understanding of snake venom composition will likely unearth novel biologically active proteins and peptides that have promising therapeutic or biotechnological applications. While a bottom-up proteomic workflow has been the main approach for cataloguing snake venom compositions at the toxin family level, it is unable to capture snake venom heterogeneity in the form of protein isoforms and higher-order protein interactions that are important in driving venom toxicity but remain underexplored. This review aims to highlight the importance of understanding snake venom heterogeneity beyond the primary sequence, in the form of post-translational modifications that give rise to different proteoforms and the myriad of higher-order protein complexes in snake venoms. We focus on current top-down proteomic workflows to identify snake venom proteoforms and further discuss alternative or novel separation, instrumentation, and data processing strategies that may improve proteoform identification. The current higher-order structural characterisation techniques implemented for snake venom proteins are also discussed; we emphasise the need for complementary and higher resolution structural bioanalytical techniques such as mass spectrometry-based approaches, X-ray crystallography and cryogenic electron microscopy, to elucidate poorly characterised tertiary and quaternary protein structures. We envisage that the expansion of the snake venom characterisation "toolbox" with top-down proteomics and high-resolution protein structure determination techniques will be pivotal in advancing structural understanding of snake venoms towards the development of improved therapeutic and biotechnology applications.
Collapse
Affiliation(s)
- C Ruth Wang
- Discipline of Chemistry, School of Physics, Chemistry and Earth Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Lewis O McFarlane
- Discipline of Chemistry, School of Physics, Chemistry and Earth Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Tara L Pukala
- Discipline of Chemistry, School of Physics, Chemistry and Earth Sciences, The University of Adelaide, Adelaide, 5005, Australia.
| |
Collapse
|
9
|
Bialves TS, Bastos LL, Parra JAA, Moysés MN, Marques E, de Castro Pimenta AM, Quintela FM, Mariano DCB, Carvalho FC, de Melo-Minardi RC, Boyle RT. Interaction of DisBa01 peptide from Bothrops alternatus venom with BRAF melanoma receptors: Modeling and molecular docking. Int J Biol Macromol 2024; 274:133283. [PMID: 38909731 DOI: 10.1016/j.ijbiomac.2024.133283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Metastatic melanoma is highly aggressive and challenging, often leading to a grim prognosis. Its progression is swift, especially when mutations like BRAFV600E continuously activate pathways vital for cell growth and survival. Although several treatments target this mutation, resistance typically emerges over time. In recent decades, research has underscored the potential of snake venoms and peptides as bioactive substances for innovative drugs, including anti-coagulants, anti-microbial, and anti-cancer agents. Leveraging this knowledge, we propose employing a bioinformatics simulation approach to: a) Predict how well a peptide (DisBa01) from Bothrops alternatus snake venom binds to the melanoma receptor BRAFV600E via Molecular Docking. b) Identify the specific peptide binding sites on receptors and analyze their proximity to active receptor sites. c) Evaluate the behavior of resulting complexes through molecular dynamics simulations. d) Assess whether this peptide qualifies as a candidate for anti-melanoma therapy. Our findings reveal that DisBa01 enhances stability in the BRAFV600E melanoma receptor structure by binding to its RGD motif, an interaction absent in the BRAF WT model. Consequently, both docking and molecular dynamics simulations suggest that DisBa01 shows promise as a BRAFV600E inhibitor.
Collapse
Affiliation(s)
- Tatiane Senna Bialves
- Graduate Program in Physiological Sciences (PPGCF), Federal University of Rio Grande - FURG, Av. Italy, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil.
| | - Luana Luiza Bastos
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - John Alexanders Amaya Parra
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maurício Nogueira Moysés
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edleusa Marques
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Graduate Program in Biochemistry and Immunology, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Marques Quintela
- Instituto Nacional de Pesquisas do Pantanal- Museu Paraense Emílio Goeldi, Av. Magalhães Barata, 376, Belém, Pará, Brazil
| | - Diego César Batista Mariano
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frederico Chaves Carvalho
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Raquel C de Melo-Minardi
- Laboratory of Bioinformatics and Systems, Institute of Exact Sciences, Department of Computer Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robert Tew Boyle
- Graduate Program in Physiological Sciences (PPGCF), Federal University of Rio Grande - FURG, Av. Italy, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil
| |
Collapse
|
10
|
Bourke LA, Zdenek CN, Huynh TM, Hodgson WC, Alagón A, Castro EN, Jones J, Fry BG. Fangs and foliage: Unearthing the haemotoxic secrets of cannabis-dwelling rattlesnakes. Toxicon 2024; 244:107756. [PMID: 38740096 DOI: 10.1016/j.toxicon.2024.107756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Despite a recent surge in high-throughput venom research that has enabled many species to be studied, some snake venoms remain understudied. The long-tailed rattlesnakes (Crotalus ericsmithi, C. lannomi, and C. stejnegeri) are one group where such research lags, largely owing to the rarity of these snakes and the hazardous areas, ripe with drug (marijuana and opium) production, they inhabit in Mexico. To fill this knowledge gap, we used multiple functional assays to examine the coagulotoxic (including across different plasma types), neurotoxic, and myotoxic activity of the venom of the long-tailed rattlesnakes. All crude venoms were shown to be potently anticoagulant on human plasma, which we discovered was not due to the destruction of fibrinogen, except for C. stejnegeri displaying minor fibrinogen destruction activity. All venoms exhibited anticoagulant activity on rat, avian, and amphibian plasmas, with C. ericsmithi being the most potent. We determined the mechanism of anticoagulant activity by C. ericsmithi and C. lannomi venoms to be phospholipid destruction and inhibition of multiple coagulation factors, leading to a net disruption of the clotting cascade. In the chick biventer assay, C. ericsmithi and C. lannomi did not exhibit neurotoxic activity but displayed potential weak myotoxic activity. BIRMEX® (Faboterápico Polivalente Antiviperino) antivenom was not effective in neutralising this venom effect. Overall, this study provides an in-depth investigation of venom function of understudied long-tailed rattlesnakes and provides a springboard for future venom and ecology research on the group.
Collapse
Affiliation(s)
- Lachlan A Bourke
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Christina N Zdenek
- School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Tam M Huynh
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Wayne C Hodgson
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Alejandro Alagón
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Av. Universidad 2001, Cuernavaca, 62210, Mexico
| | - Edgar N Castro
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Av. Universidad 2001, Cuernavaca, 62210, Mexico; Investigador por México, Facultad de Ciencias Biológicas, Universidad Juárez del Estado de Durango, Avenida Universidad s/n. Fracc. Filadelfia, C.P. 35010, Gómez Palacio, Dgo., Mexico
| | - Jason Jones
- Herp.mx A.C, Villa Del Álvarez, Colima, Mexico
| | - Bryan G Fry
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
11
|
Hsiao MH, Miao Y, Liu Z, Schütze K, Limjunyawong N, Chien DCC, Monteiro WD, Chu LS, Morgenlander W, Jayaraman S, Jang SE, Gray JJ, Zhu H, Dong X, Steinegger M, Larman HB. Molecular Display of the Animal Meta-Venome for Discovery of Novel Therapeutic Peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.595990. [PMID: 38854075 PMCID: PMC11160688 DOI: 10.1101/2024.05.27.595990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Animal venoms, distinguished by their unique structural features and potent bioactivities, represent a vast and relatively untapped reservoir of therapeutic molecules. However, limitations associated with extracting or expressing large numbers of individual venoms and venom-like molecules have precluded their therapeutic evaluation via high throughput screening. Here, we developed an innovative computational approach to design a highly diverse library of animal venoms and "metavenoms". We employed programmable M13 hyperphage display to preserve critical disulfide-bonded structures for highly parallelized single-round biopanning with quantitation via high-throughput DNA sequencing. Our approach led to the discovery of Kunitz type domain containing proteins that target the human itch receptor Mas-related G protein-coupled receptor X4 (MRGPRX4), which plays a crucial role in itch perception. Deep learning-based structural homology mining identified two endogenous human homologs, tissue factor pathway inhibitor (TFPI) and serine peptidase inhibitor, Kunitz type 2 (SPINT2), which exhibit agonist-dependent potentiation of MRGPRX4. Highly multiplexed screening of animal venoms and metavenoms is therefore a promising approach to uncover new drug candidates.
Collapse
Affiliation(s)
- Meng-Hsuan Hsiao
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- These authors contributed equally to this work
| | - Yang Miao
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- These authors contributed equally to this work
| | - Zixing Liu
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biology, Zanvyl Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Konstantin Schütze
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center of Research Excellence in Allergy and Immunology, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Daphne Chun-Che Chien
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wayne Denis Monteiro
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lee-Shin Chu
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - William Morgenlander
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sahana Jayaraman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sung-eun Jang
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jeffrey J. Gray
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Viral Oncology Program, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Martin Steinegger
- School of Biological Sciences, Seoul National University, Seoul, South Korea
- Artificial Intelligence Institute, Seoul National University, Seoul, South Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Shahdadi S, Hamidi F, Fathi B. The effect of Iranian snake, Naja naja oxiana venom on the blood glucose concentration and some biochemical parameters of experimental diabetic rats. Heliyon 2024; 10:e24436. [PMID: 38288016 PMCID: PMC10823082 DOI: 10.1016/j.heliyon.2024.e24436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/31/2024] Open
Abstract
Diabetes is a chronic disease resulting from impaired insulin production and function; leading to hyperglycaemia and long-term complications. The treatment for Type I diabetes treatment involves insulin injections while Type II diabetes treatments include drugs such as metformin and sulfonylureas, along with lifestyle changes. These medicines can be expensive and may have adverse effects. Therefore, the search for new therapeutic agents continues. Venoms from various animals yield numerous pharmacologically active compounds. In this study, we investigated the effects of the venom from an Iranian snake, Naja naja oxiana, on blood glucose concentration and certain serum biochemical parameters in male rats with induced diabetes. Diabetes was induced in male rats using either a single injection of streptozotocin (STZ) alone (55 mg/kg i. p.) or STZ (65 mg/kg i. p.) preceded by nicotinamide (230/kg i. p.) administered 15 min earlier. The diabetic rats produced by either method received a single injection of either vehicle or venom (0.2 or 0.4 mg/kg i. p.). In the STZ rats, this was done 13 days after diabetes induction, while in the STZ-nicotinamide rats, venom was injected 3 days after diabetes induction. The venom from Naja naja oxiana significantly reduced blood glucose levels in male rats with diabetes induced by either method. Additionally, the venom decreased serum cholesterol and triglycerides concentrations. However, the venom had no effect on the blood glucose levels of healthy male rats. Pretreatment with the venom did not prevent the induction of diabetes by STZ. These findings suggest that Naja naja oxiana venom exhibits an anti-diabetic effect and could be a potential candidate for effectively controlling diabetes.
Collapse
Affiliation(s)
- Shiva Shahdadi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Farshid Hamidi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Behrooz Fathi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
13
|
Ibrahim MAA, Abdeljawaad KAA, Abdelrahman AHM, Sidhom PA, Tawfeek AM, Mekhemer GAH, Abd El-Rahman MK, Dabbish E, Shoeib T. In-Silico Mining of the Toxins Database (T3DB) towards Hunting Prospective Candidates as ABCB1 Inhibitors: Integrated Molecular Docking and Lipid Bilayer-Enhanced Molecular Dynamics Study. Pharmaceuticals (Basel) 2023; 16:1019. [PMID: 37513931 PMCID: PMC10384459 DOI: 10.3390/ph16071019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Multidrug resistance (MDR) is one of the most problematic issues in chemotherapeutic carcinoma therapy. The ABCB1 transporter, a drug efflux pump overexpressed in cancer cells, has been thoroughly investigated for its association with MDR. Thus, discovering ABCB1 inhibitors can reverse the MDR in cancer cells. In the current work, a molecular docking technique was utilized for hunting the most prospective ABCB1 inhibitors from the Toxin and Toxin-Target Database (T3DB). Based on the docking computations, the most promising T3DB compounds complexed with the ABCB1 transporter were subjected to molecular dynamics (MD) simulations over 100 ns. Utilizing the MM-GBSA approach, the corresponding binding affinities were computed. Compared to ZQU (calc. -49.8 kcal/mol), Emamectin B1a (T3D1043), Emamectin B1b (T3D1044), Vincristine (T3D4016), Vinblastine (T3D4017), and Vindesine (T3D2479) complexed with ABCB1 transporter demonstrated outstanding binding affinities with ΔGbinding values of -93.0, -92.6, -93.8, -92.2, and -90.8 kcal/mol, respectively. The structural and energetic investigations confirmed the constancy of the identified T3DB compounds complexed with the ABCB1 transporter during the 100 ns MD course. To mimic the physiological conditions, MD simulations were conducted for those identified inhibitors complexed with ABCB1 transporter in the presence of a POPC membrane. These findings revealed that Emamectin B1a, Emamectin B1b, Vincristine, Vinblastine, and Vindesine are promising ABCB1 inhibitors that can reverse the MDR. Therefore, subjecting those compounds to further in-vitro and in-vivo investigations is worthwhile.
Collapse
Affiliation(s)
- Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
- School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Khlood A A Abdeljawaad
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Ahmed M Tawfeek
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Gamal A H Mekhemer
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Mohamed K Abd El-Rahman
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Eslam Dabbish
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Tamer Shoeib
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
14
|
Gable SM, Mendez JM, Bushroe NA, Wilson A, Byars MI, Tollis M. The State of Squamate Genomics: Past, Present, and Future of Genome Research in the Most Speciose Terrestrial Vertebrate Order. Genes (Basel) 2023; 14:1387. [PMID: 37510292 PMCID: PMC10379679 DOI: 10.3390/genes14071387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Squamates include more than 11,000 extant species of lizards, snakes, and amphisbaenians, and display a dazzling diversity of phenotypes across their over 200-million-year evolutionary history on Earth. Here, we introduce and define squamates (Order Squamata) and review the history and promise of genomic investigations into the patterns and processes governing squamate evolution, given recent technological advances in DNA sequencing, genome assembly, and evolutionary analysis. We survey the most recently available whole genome assemblies for squamates, including the taxonomic distribution of available squamate genomes, and assess their quality metrics and usefulness for research. We then focus on disagreements in squamate phylogenetic inference, how methods of high-throughput phylogenomics affect these inferences, and demonstrate the promise of whole genomes to settle or sustain persistent phylogenetic arguments for squamates. We review the role transposable elements play in vertebrate evolution, methods of transposable element annotation and analysis, and further demonstrate that through the understanding of the diversity, abundance, and activity of transposable elements in squamate genomes, squamates can be an ideal model for the evolution of genome size and structure in vertebrates. We discuss how squamate genomes can contribute to other areas of biological research such as venom systems, studies of phenotypic evolution, and sex determination. Because they represent more than 30% of the living species of amniote, squamates deserve a genome consortium on par with recent efforts for other amniotes (i.e., mammals and birds) that aim to sequence most of the extant families in a clade.
Collapse
Affiliation(s)
- Simone M Gable
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Jasmine M Mendez
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Nicholas A Bushroe
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Adam Wilson
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Michael I Byars
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Marc Tollis
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| |
Collapse
|
15
|
Ferreira PMP, Sousa LQD, Sousa RWRD, Rodrigues DDJ, Monção Filho EDS, Chaves MH, Vieira Júnior GM, Rizzo MDS, Filgueiras LA, Mendes AN, Lima DJB, Pessoa C, Sousa JMDCE, Rodrigues ACBDC, Soares MBP, Bezerra DP. Toxic profile of marinobufagin from poisonous Amazon toads and antitumoral effects on human colorectal carcinomas. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116406. [PMID: 36965547 DOI: 10.1016/j.jep.2023.116406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/10/2023] [Accepted: 03/18/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE South Americans natives have extensively used the toad "kururu" to reduce/treat skin infections, cutaneous lesions and sores. They release secretions rich in bufadienolides, polyhydroxy steroids with well-documented cardiotonic and antiproliferative actions, but in vivo antitumoral evaluations in mammals are rare, and toxicological safety has been left in second place. AIMS OF THE STUDY This investigation used in silico, in vitro and in vivo tools to evaluate acute and subacute toxic effects of marinobufagin and the anticancer action in tumor-bearing mice models. MATERIALS AND METHODS Initially, in silico toxic predictions were performed, followed by in vitro assays using human and murine normal and tumor lines. Next, acute and subacute studies on mice investigated the behavior, hematological and intestinal transit profile and antitumoral activity of marinobufagin in sarcoma 180- and HCT-116 colorectal carcinoma-transplanted mice for 7 and 15 days, respectively. Ex vivo and in vivo cytogenetic assays in Sarcoma 180 and bone marrow cells and histopathological examinations were also executed. RESULTS In silico studies revealed ecotoxicological effects on crustaceans (Daphnia sp.), fishes (Pimephales promelas and Oryzias latipes), and algae. A 24-h marinobufagin-induced acute toxicity included signals of central activity, mainly (vocal frenzy, absence of body tonus, increased ventilation, ataxia, and equilibrium loss), and convulsions and death at 10 mg/kg. The bufadienolide presented effective in vitro cytotoxic action on human lines of colorectal carcinomas in a similar way to ouabain and tumor reduction in marinobufagin-treated SCID-bearing HCT-116 heterotopic xenografts. Animals under subacute nonlethal doses exhibited a decrease in creatinine clearance with normal levels of blood urea, probably as a result of a marinobufagin-induced renal perfusion fall. Nevertheless, only minor morphological side effects were identified in kidneys, livers, hearts and lungs. CONCLUSIONS Marinobufagin has in vitro and in vivo anticancer action on colorectal carcinoma and mild and reversible alterations in key metabolic organs without direct chemotherapy-induced gastrointestinal effects at subacute exposure, but it causes acute ataxia, equilibrium loss, convulsions and death at higher acute exposure.
Collapse
Affiliation(s)
- Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, 64049-550, Teresina, Brazil.
| | - Lívia Queiroz de Sousa
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, 64049-550, Teresina, Brazil
| | - Rayran Walter Ramos de Sousa
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, 64049-550, Teresina, Brazil
| | - Domingos de Jesus Rodrigues
- Institute of Natural, Humanities and Social Sciences, Federal University of Mato Grosso, 78550-728, Sinop, Brazil
| | | | - Mariana Helena Chaves
- Laboratory of Natural Products, Department of Chemistry, Federal University of Piauí, 64049-550, Teresina, Brazil
| | - Gerardo Magela Vieira Júnior
- Laboratory of Natural Products, Department of Chemistry, Federal University of Piauí, 64049-550, Teresina, Brazil
| | | | - Lívia Alves Filgueiras
- Laboratory of Innovation in Science and Technology (Lacitec), Department of Biophysics and Physiology, Federal University of Piauí, 64049-550, Teresina, Brazil
| | - Anderson Nogueira Mendes
- Laboratory of Innovation in Science and Technology (Lacitec), Department of Biophysics and Physiology, Federal University of Piauí, 64049-550, Teresina, Brazil
| | - Daisy Jereissati Barbosa Lima
- Laboratory of Experimental Oncology (LOE), Department of Physiology and Pharmacology, Federal University of Ceará, 60430-270, Fortaleza, Brazil
| | - Cláudia Pessoa
- Laboratory of Experimental Oncology (LOE), Department of Physiology and Pharmacology, Federal University of Ceará, 60430-270, Fortaleza, Brazil
| | - João Marcelo de Castro E Sousa
- Laboraroty of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, 64049-550, Teresina, Brazil
| | | | | | - Daniel Pereira Bezerra
- Laboratory of Tissue Engineering and Immunopharmacology, Oswaldo Cruz Foundation, 40296-710, Salvador, Brazil
| |
Collapse
|
16
|
David V, Wermelinger LS, Frattani FS, Lima AGF, Santos YFS, Mourão PADS, Almeida FCL, Kurtenbach E, Zingali RB. rJararacin, a recombinant disintegrin from Bothrops jararaca venom: Exploring its effects on hemostasis and thrombosis. Arch Biochem Biophys 2023; 738:109557. [PMID: 36878339 DOI: 10.1016/j.abb.2023.109557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Integrins are a family of heterodimeric transmembrane receptors which link the extracellular matrix to the cell cytoskeleton. These receptors play a role in many cellular processes: adhesion, proliferation, migration, apoptosis, and platelet aggregation, thus modulating a wide range of scenarios in health and disease. Therefore, integrins have been the target of new antithrombotic drugs. Disintegrins from snake venoms are recognized by the ability to modulate the activity of integrins, such as integrin αIIbβ3, a fundamental platelet glycoprotein, and αvβ3 expressed on tumor cells. For this reason, disintegrins are unique and potential tools for examining integrin-matrix interaction and the development of novel antithrombotic agents. The present study aims to obtain the recombinant form of jararacin and evaluate the secondary structure and its effects on hemostasis and thrombosis. rJararacin was expressed in the Pichia pastoris (P. pastoris) expression system and purified the recombinant protein with a yield of 40 mg/L of culture. The molecular mass (7722 Da) and internal sequence were confirmed by mass spectrometry. Structure and folding analysis were obtained by Circular Dichroism and 1H Nuclear Magnetic Resonance spectra. Disintegrin structure reveals properly folded with the presence of β-sheet structure. rJararacin significantly demonstrated inhibition of the adhesion of B16F10 cells and platelets to the fibronectin matrix under static conditions. rJararacin inhibited platelet aggregation induced by ADP (IC50 95 nM), collagen (IC50 57 nM), and thrombin (IC50 22 nM) in a dose-dependent manner. This disintegrin also inhibited 81% and 94% of the adhesion of platelets to fibrinogen and collagen under continuous flow, respectively. In addition, rjararacin efficaciously prevents platelet aggregation in vitro and ex vivo with rat platelets and thrombus occlusion at an effective dose (5 mg/kg). The data here provides evidence that rjararacin possesses the potential as an αIIbβ3 antagonist, capable of preventing arterial thrombosis.
Collapse
Affiliation(s)
- Victor David
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Luciana Serrão Wermelinger
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-170, Brazil.
| | - Flávia Serra Frattani
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-170, Brazil.
| | - Antonio Gilclêr Ferreira Lima
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Yasmyn Fernandes Silva Santos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Paulo Antônio de Souza Mourão
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Fabio Ceneviva Lacerda Almeida
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| | - Eleonora Kurtenbach
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-170, Brazil.
| | - Russolina Benedeta Zingali
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, CEP 21941-902, Brazil.
| |
Collapse
|
17
|
Bialves TS, Bastos Junior CLQ, Cordeiro MF, Boyle RT. Snake venom, a potential treatment for melanoma. A systematic review. Int J Biol Macromol 2023; 231:123367. [PMID: 36690229 DOI: 10.1016/j.ijbiomac.2023.123367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Despite advances in treating patients with melanoma, there are still many treatment challenges to overcome. Studies with snake venom-derived proteins/peptides describe their binding potential, and inhibition of some proliferative mechanisms in melanoma. The combined use of these compounds with current therapies could be the strategic gap that will help us discover more effective treatments for melanoma. The present study aimed to carry out a systematic review identifying snake venom proteins and peptides described in the literature with antitumor, antimetastatic, or antiangiogenic effects on melanoma and determine the mechanisms of action that lead to these anti-tumor effects. Snake venoms contain proteins and peptides which are antiaggregant, antimetastatic, and antiangiogenic. The in vivo results are encouraging, considering the reduction of metastases and tumor size after treatment. In addition to these results, it was reported that these venom compounds could act in combination with chemotherapeutics (Acurhagin-C; Macrovipecetin), sensitizing and preparing tumor cells for treatment. There is a consensus that snake venom is a promising strategy for the improvement of antimelanoma therapies, but it has been little explored in the current context, combined with inhibitors, immunotherapy or tumor microenvironment, for example. We suggest Lebein as a candidate for combination therapy with BRAF inhibitors.
Collapse
Affiliation(s)
- Tatiane Senna Bialves
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil.
| | - Claudio L Q Bastos Junior
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil
| | - Marcos Freitas Cordeiro
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Universidade do Oeste de Santa Catarina - UNOESC, Rua Roberto Trompovski 224, Joaçaba, Santa Catarina, CEP 89600-000, Brazil.
| | - Robert Tew Boyle
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil; Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, Rio Grande do Sul 96203-900, Brazil
| |
Collapse
|
18
|
Bioactive peptides from scorpion venoms: therapeutic scaffolds and pharmacological tools. Chin J Nat Med 2023; 21:19-35. [PMID: 36641229 DOI: 10.1016/s1875-5364(23)60382-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 01/14/2023]
Abstract
Evolution and natural selection have endowed animal venoms, including scorpion venoms, with a wide range of pharmacological properties. Consequently, scorpions, their venoms, and/or their body parts have been used since time immemorial in traditional medicines, especially in Africa and Asia. With respect to their pharmacological potential, bioactive peptides from scorpion venoms have become an important source of scientific research. With the rapid increase in the characterization of various components from scorpion venoms, a large number of peptides are identified with an aim of combating a myriad of emerging global health problems. Moreover, some scorpion venom-derived peptides have been established as potential scaffolds helpful for drug development. In this review, we summarize the promising scorpion venoms-derived peptides as drug candidates. Accordingly, we highlight the data and knowledge needed for continuous characterization and development of additional natural peptides from scorpion venoms, as potential drugs that can treat related diseases.
Collapse
|
19
|
The First Snake Venom KTS/Disintegrins-Integrin Interactions Using Bioinformatics Approaches. Molecules 2022; 28:molecules28010325. [PMID: 36615520 PMCID: PMC9822126 DOI: 10.3390/molecules28010325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/26/2022] [Accepted: 12/09/2022] [Indexed: 01/03/2023] Open
Abstract
Snake venom contains a number of active molecules that have been shown to possess high anti-tumor activities; disintegrins are an excellent example among these. Their ability to interact and bind with integrins suggests that they could be very valuable molecules for the development of new cancer therapeutic approaches. However, in the absence of a clear Lysine-Threonine-Serine (KTS) Disintegrins Integrin interaction model, the exact compound features behind it are still unknown. In this study, we investigated the structural characteristics of three KTS-disintegrins and the interaction mechanisms with the α1β1 integrin receptor using in silico bioinformatics approaches. Normal mode analysis showed that the flexibility of the KTSR motif and the C-terminal region play a key role and influence the KTS-Disintegrin-integrin interaction. Protein-protein docking also suggested that the interaction involving the KTSR motif is highly dependent on the residue following K21, S23 and R24. These findings contribute to a better understanding of the KTS-Disintegrin-Integrin structural differences and their interactions with α1β1 receptors, which could improve the selection process of the best active molecules for antitumor therapies.
Collapse
|
20
|
Wells M, Fossépré M, Hambye S, Surin M, Blankert B. Uncovering the antimalarial potential of toad venoms through a bioassay-guided fractionation process. Int J Parasitol Drugs Drug Resist 2022; 20:97-107. [PMID: 36343571 PMCID: PMC9772263 DOI: 10.1016/j.ijpddr.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/16/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Malaria remains to date one of the most devastating parasitic diseases worldwide. The fight against this disease is rendered more difficult by the emergence and spread of drug-resistant strains. The need for new therapeutic candidates is now greater than ever. In this study, we investigated the antiplasmodial potential of toad venoms. The wide array of bioactive compounds present in Bufonidae venoms has allowed researchers to consider many potential therapeutic applications, especially for cancers and infectious diseases. We focused on small molecules, namely bufadienolides, found in the venom of Rhinella marina (L.). The developed bio-guided fractionation process includes a four solvent-system extraction followed by fractionation using flash chromatography. Sub-fractions were obtained through preparative TLC. All samples were characterized using chromatographic and spectrometric techniques and then underwent testing on in vitro Plasmodium falciparum cultures. Two strains were considered: 3D7 (chloroquine-sensitive) and W2 (chloroquine-resistant). This strategy highlighted a promising activity for one compound named resibufogenin. With IC50 values of (29 ± 8) μg/mL and (23 ± 1) μg/mL for 3D7 and W2 respectively, this makes it an interesting candidate for further investigation. A molecular modelling approach proposed a potential binding mode of resibufogenin to Plasmodium falciparum adenine-triphosphate 4 pump as antimalarial drug target.
Collapse
Affiliation(s)
- Mathilde Wells
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons - UMONS, Place du Parc 20, 7000, Mons, Belgium
| | - Mathieu Fossépré
- Laboratory for Chemistry of Novel Materials, Faculty of Sciences, Research Institute for Biosciences and Research Institute for Materials, University of Mons - UMONS, Place du Parc 20, 7000, Mons, Belgium
| | - Stéphanie Hambye
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons - UMONS, Place du Parc 20, 7000, Mons, Belgium
| | - Mathieu Surin
- Laboratory for Chemistry of Novel Materials, Faculty of Sciences, Research Institute for Biosciences and Research Institute for Materials, University of Mons - UMONS, Place du Parc 20, 7000, Mons, Belgium
| | - Bertrand Blankert
- Laboratory of Pharmaceutical Analysis, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons - UMONS, Place du Parc 20, 7000, Mons, Belgium.
| |
Collapse
|
21
|
Seaweeds in the Oncology Arena: Anti-Cancer Potential of Fucoidan as a Drug—A Review. Molecules 2022; 27:molecules27186032. [PMID: 36144768 PMCID: PMC9506145 DOI: 10.3390/molecules27186032] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Marine natural products are a discerning arena to search for the future generation of medications to treat a spectrum of ailments. Meanwhile, cancer is becoming more ubiquitous over the world, and the likelihood of dying from it is rising. Surgery, radiation, and chemotherapy are the mainstays of cancer treatment worldwide, but their extensive side effects limit their curative effect. The quest for low-toxicity marine drugs to prevent and treat cancer is one of the current research priorities of researchers. Fucoidan, an algal sulfated polysaccharide, is a potent therapeutic lead candidate against cancer, signifying that far more research is needed. Fucoidan is a versatile, nontoxic marine-origin heteropolysaccharide that has received much attention due to its beneficial biological properties and safety. Fucoidan has been demonstrated to exhibit a variety of conventional bioactivities, such as antiviral, antioxidant, and immune-modulatory characteristics, and anticancer activity against a wide range of malignancies has also recently been discovered. Fucoidan inhibits tumorigenesis by prompting cell cycle arrest and apoptosis, blocking metastasis and angiogenesis, and modulating physiological signaling molecules. This review compiles the molecular and cellular aspects, immunomodulatory and anticancer actions of fucoidan as a natural marine anticancer agent. Specific fucoidan and membranaceous polysaccharides from Ecklonia cava, Laminaria japonica, Fucus vesiculosus, Astragalus, Ascophyllum nodosum, Codium fragile serving as potential anticancer marine drugs are discussed in this review.
Collapse
|
22
|
Repositioning of tubocurarine as analgesic and anti-inflammatory agent: Exploring beyond myorelaxant activity. Biochem Pharmacol 2022; 205:115248. [PMID: 36113566 DOI: 10.1016/j.bcp.2022.115248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/03/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND PURPOSE Tubocurarine (d-TC), a non-depolarizing competitive blocker of nicotinic acetylcholine receptors is extensively utilized for the relaxation of skeletal muscles. Drug repositioning is a forthright approach to reduce the cost and speed up drug development process. Herein, we have attempted to evaluate the analgesic and anti-inflammatory activity of d-TC for its possible repurposing in pain and inflammation-related issues. EXPERIMENTAL APPROACH We examined the soluble epoxide hydrolase inhibitory (sEHI) activity of d-TC employing in silico high throughput screening protocols, in vitro cell-free sEH inhibitory assay, and in in vivo rodent models for its repositioning in pain and inflammation-related disorders. KEY RESULTS In molecular docking study, d-TC displayed impressive hydrogen bonding interactions within the cavity of sEH enzyme with good docking score. d-TC also exhibited notable sEH inhibitory activity (IC50 3.72 nm) at the in vitro assay. Oral absorption capability of d-TC (0.1 and 0.2 mg/mL) was determined using an in vitro everted intestinal sac model employing rat ileum tissue that revealed significant oral absorption of d-TC. Besides, in vivo studies revealed that oral administration of d-TC (0.1 and 0.2 mg/kg) in rodents significantly attenuated hyperalgesia (cold plate test, tail immersion test and formalin test) and inflammation (estimation of rectal temperature, acetic acid induced pleurisy test and cotton pellet-induced granuloma test) induced in robust preclinical models. Conclusion and implications These findings are novel and warrant immediate efforts to reposition d-TC as a new therapeutic candidate in the management of hyperalgesia, inflammation, and associated conditions.
Collapse
|
23
|
von Reumont BM, Anderluh G, Antunes A, Ayvazyan N, Beis D, Caliskan F, Crnković A, Damm M, Dutertre S, Ellgaard L, Gajski G, German H, Halassy B, Hempel BF, Hucho T, Igci N, Ikonomopoulou MP, Karbat I, Klapa MI, Koludarov I, Kool J, Lüddecke T, Ben Mansour R, Vittoria Modica M, Moran Y, Nalbantsoy A, Ibáñez MEP, Panagiotopoulos A, Reuveny E, Céspedes JS, Sombke A, Surm JM, Undheim EAB, Verdes A, Zancolli G. Modern venomics-Current insights, novel methods, and future perspectives in biological and applied animal venom research. Gigascience 2022; 11:giac048. [PMID: 35640874 PMCID: PMC9155608 DOI: 10.1093/gigascience/giac048] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022] Open
Abstract
Venoms have evolved >100 times in all major animal groups, and their components, known as toxins, have been fine-tuned over millions of years into highly effective biochemical weapons. There are many outstanding questions on the evolution of toxin arsenals, such as how venom genes originate, how venom contributes to the fitness of venomous species, and which modifications at the genomic, transcriptomic, and protein level drive their evolution. These questions have received particularly little attention outside of snakes, cone snails, spiders, and scorpions. Venom compounds have further become a source of inspiration for translational research using their diverse bioactivities for various applications. We highlight here recent advances and new strategies in modern venomics and discuss how recent technological innovations and multi-omic methods dramatically improve research on venomous animals. The study of genomes and their modifications through CRISPR and knockdown technologies will increase our understanding of how toxins evolve and which functions they have in the different ontogenetic stages during the development of venomous animals. Mass spectrometry imaging combined with spatial transcriptomics, in situ hybridization techniques, and modern computer tomography gives us further insights into the spatial distribution of toxins in the venom system and the function of the venom apparatus. All these evolutionary and biological insights contribute to more efficiently identify venom compounds, which can then be synthesized or produced in adapted expression systems to test their bioactivity. Finally, we critically discuss recent agrochemical, pharmaceutical, therapeutic, and diagnostic (so-called translational) aspects of venoms from which humans benefit.
Collapse
Affiliation(s)
- Bjoern M von Reumont
- Goethe University Frankfurt, Institute for Cell Biology and Neuroscience, Department for Applied Bioinformatics, 60438 Frankfurt am Main, Germany
- LOEWE Centre for Translational Biodiversity Genomics, Senckenberg Frankfurt, Senckenberganlage 25, 60235 Frankfurt, Germany
- Justus Liebig University Giessen, Institute for Insectbiotechnology, Heinrich Buff Ring 26-32, 35396 Giessen, Germany
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Agostinho Antunes
- CIIMAR/CIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
| | - Naira Ayvazyan
- Orbeli Institute of Physiology of NAS RA, Orbeli ave. 22, 0028 Yerevan, Armenia
| | - Dimitris Beis
- Developmental Biology, Centre for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Figen Caliskan
- Department of Biology, Faculty of Science and Letters, Eskisehir Osmangazi University, TR-26040 Eskisehir, Turkey
| | - Ana Crnković
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Maik Damm
- Technische Universität Berlin, Department of Chemistry, Straße des 17. Juni 135, 10623 Berlin, Germany
| | | | - Lars Ellgaard
- Department of Biology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Goran Gajski
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska cesta 2, 10000 Zagreb, Croatia
| | - Hannah German
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Beata Halassy
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Trg Republike Hrvatske 14, 10000 Zagreb, Croatia
| | - Benjamin-Florian Hempel
- BIH Center for Regenerative Therapies BCRT, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Nasit Igci
- Nevsehir Haci Bektas Veli University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, 50300 Nevsehir, Turkey
| | - Maria P Ikonomopoulou
- Madrid Institute for Advanced Studies in Food, Madrid,E28049, Spain
- The University of Queensland, St Lucia, QLD 4072, Australia
| | - Izhar Karbat
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Maria I Klapa
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research & Technology Hellas (FORTH/ICE-HT), Patras GR-26504, Greece
| | - Ivan Koludarov
- Justus Liebig University Giessen, Institute for Insectbiotechnology, Heinrich Buff Ring 26-32, 35396 Giessen, Germany
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Tim Lüddecke
- LOEWE Centre for Translational Biodiversity Genomics, Senckenberg Frankfurt, Senckenberganlage 25, 60235 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, 35392 Gießen, Germany
| | - Riadh Ben Mansour
- Department of Life Sciences, Faculty of Sciences, Gafsa University, Campus Universitaire Siidi Ahmed Zarrouk, 2112 Gafsa, Tunisia
| | - Maria Vittoria Modica
- Dept. of Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Via Po 25c, I-00198 Roma, Italy
| | - Yehu Moran
- Department of Ecology, Evolution and Behavior, Alexander Silberman Institute of Life Sciences, Faculty of Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ayse Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Bornova, Izmir, Turkey
| | - María Eugenia Pachón Ibáñez
- Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Virgen del Rocío University Hospital, Institute of Biomedicine of Seville, 41013 Sevilla, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alexios Panagiotopoulos
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research & Technology Hellas (FORTH/ICE-HT), Patras GR-26504, Greece
- Animal Biology Division, Department of Biology, University of Patras, Patras, GR-26500, Greece
| | - Eitan Reuveny
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Javier Sánchez Céspedes
- Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Virgen del Rocío University Hospital, Institute of Biomedicine of Seville, 41013 Sevilla, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Andy Sombke
- Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Joachim M Surm
- Department of Ecology, Evolution and Behavior, Alexander Silberman Institute of Life Sciences, Faculty of Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Eivind A B Undheim
- University of Oslo, Centre for Ecological and Evolutionary Synthesis, Postboks 1066 Blindern 0316 Oslo, Norway
| | - Aida Verdes
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, José Gutiérrez Abascal 2, 28006 Madrid, Spain
| | - Giulia Zancolli
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| |
Collapse
|
24
|
Exploring Toxins for Hunting SARS-CoV-2 Main Protease Inhibitors: Molecular Docking, Molecular Dynamics, Pharmacokinetic Properties, and Reactome Study. Pharmaceuticals (Basel) 2022; 15:ph15020153. [PMID: 35215266 PMCID: PMC8875976 DOI: 10.3390/ph15020153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 12/13/2022] Open
Abstract
The main protease (Mpro) is a potential druggable target in SARS-CoV-2 replication. Herein, an in silico study was conducted to mine for Mpro inhibitors from toxin sources. A toxin and toxin-target database (T3DB) was virtually screened for inhibitor activity towards the Mpro enzyme utilizing molecular docking calculations. Promising toxins were subsequently characterized using a combination of molecular dynamics (MD) simulations and molecular mechanics-generalized Born surface area (MM-GBSA) binding energy estimations. According to the MM-GBSA binding energies over 200 ns MD simulations, three toxins—namely philanthotoxin (T3D2489), azaspiracid (T3D2672), and taziprinone (T3D2378)—demonstrated higher binding affinities against SARS-CoV-2 Mpro than the co-crystalized inhibitor XF7 with MM-GBSA binding energies of −58.9, −55.9, −50.1, and −43.7 kcal/mol, respectively. The molecular network analyses showed that philanthotoxin provides a ligand lead using the STRING database, which includes the biochemical top 20 signaling genes CTSB, CTSL, and CTSK. Ultimately, pathway enrichment analysis (PEA) and Reactome mining results revealed that philanthotoxin could prevent severe lung injury in COVID-19 patients through the remodeling of interleukins (IL-4 and IL-13) and the matrix metalloproteinases (MMPs). These findings have identified that philanthotoxin—a venom of the Egyptian solitary wasp—holds promise as a potential Mpro inhibitor and warrants further in vitro/in vivo validation.
Collapse
|
25
|
Barth T, Silva A, Santos SSD, Santos JL, Andrade PD, Tsai J, Caldas ED, Castro MDS, Pires Júnior OR. Antimicrobial activity and partial chemical structure of acylpolyamines isolated from the venom of the spider Acanthoscurria natalensis. J Venom Anim Toxins Incl Trop Dis 2022; 28:e20210017. [PMID: 35382112 PMCID: PMC8939072 DOI: 10.1590/1678-9199-jvatitd-2021-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/17/2021] [Indexed: 11/22/2022] Open
Abstract
Background: Methods: Results: Conclusion:
Collapse
|
26
|
Trevisan G, Oliveira SM. Animal Venom Peptides Cause Antinociceptive Effects by Voltage-gated Calcium Channels Activity Blockage. Curr Neuropharmacol 2022; 20:1579-1599. [PMID: 34259147 PMCID: PMC9881091 DOI: 10.2174/1570159x19666210713121217] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022] Open
Abstract
Pain is a complex phenomenon that is usually unpleasant and aversive. It can range widely in intensity, quality, and duration and has diverse pathophysiologic mechanisms and meanings. Voltage-gated sodium and calcium channels are essential to transmitting painful stimuli from the periphery until the dorsal horn of the spinal cord. Thus, blocking voltage-gated calcium channels (VGCCs) can effectively control pain refractory to treatments currently used in the clinic, such as cancer and neuropathic pain. VGCCs blockers isolated of cobra Naja naja kaouthia (α-cobratoxin), spider Agelenopsis aperta (ω-Agatoxin IVA), spider Phoneutria nigriventer (PhTx3.3, PhTx3.4, PhTx3.5, PhTx3.6), spider Hysterocrates gigas (SNX-482), cone snails Conus geographus (GVIA), Conus magus (MVIIA or ziconotide), Conus catus (CVID, CVIE and CVIF), Conus striatus (SO- 3), Conus fulmen (FVIA), Conus moncuri (MoVIA and MoVIB), Conus regularis (RsXXIVA), Conus eburneus (Eu1.6), Conus victoriae (Vc1.1.), Conus regius (RgIA), and spider Ornithoctonus huwena (huwentoxin-I and huwentoxin-XVI) venoms caused antinociceptive effects in different acute and chronic pain models. Currently, ziconotide is the only clinical used N-type VGCCs blocker peptide for chronic intractable pain. However, ziconotide causes different adverse effects, and the intrathecal route of administration also impairs its use in a more significant number of patients. In this sense, peptides isolated from animal venoms or their synthetic forms that act by modulating or blocking VGCCs channels seem to be a relevant prototype for developing new analgesics efficacious and well tolerated by patients.
Collapse
Affiliation(s)
- Gabriela Trevisan
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil
| | - Sara Marchesan Oliveira
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil
| |
Collapse
|
27
|
Hayashi MAF, Campeiro JD, Yonamine CM. Revisiting the potential of South American rattlesnake Crotalus durissus terrificus toxins as therapeutic, theranostic and/or biotechnological agents. Toxicon 2021; 206:1-13. [PMID: 34896407 DOI: 10.1016/j.toxicon.2021.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/10/2021] [Accepted: 12/08/2021] [Indexed: 02/07/2023]
Abstract
The potential biotechnological and biomedical applications of the animal venom components are widely recognized. Indeed, many components have been used either as drugs or as templates/prototypes for the development of innovative pharmaceutical drugs, among which many are still used for the treatment of human diseases. A specific South American rattlesnake, named Crotalus durissus terrificus, shows a venom composition relatively simpler compared to any viper or other snake species belonging to the Crotalus genus, although presenting a set of toxins with high potential for the treatment of several still unmet human therapeutic needs, as reviewed in this work. In addition to the main toxin named crotoxin, which is under clinical trials studies for antitumoral therapy and which has also anti-inflammatory and immunosuppressive activities, other toxins from the C. d. terrificus venom are also being studied, aiming for a wide variety of therapeutic applications, including as antinociceptive, anti-inflammatory, antimicrobial, antifungal, antitumoral or antiparasitic agent, or as modulator of animal metabolism, fibrin sealant (fibrin glue), gene carrier or theranostic agent. Among these rattlesnake toxins, the most relevant, considering the potential clinical applications, are crotamine, crotalphine and gyroxin. In this narrative revision, we propose to organize and present briefly the updates in the accumulated knowledge on potential therapeutic applications of toxins collectively found exclusively in the venom of this specific South American rattlesnake, with the objective of contributing to increase the chances of success in the discovery of drugs based on toxins.
Collapse
Affiliation(s)
- Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), SP, Brazil.
| | - Joana D Campeiro
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), SP, Brazil
| | - Camila M Yonamine
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), SP, Brazil.
| |
Collapse
|
28
|
Setayesh-Mehr Z, Poorsargol M. HL-7 and HL-10 Peptides Stimulate Insulin Secretion in the INS-1 Insulinoma Cell Line through Incretin-Dependent Pathway and Increasing the Glucose Uptake in L6 Myoblast. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10249-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Vonk FJ, Bittenbinder MA, Kerkkamp HMI, Grashof DGB, Archer JP, Afonso S, Richardson MK, Kool J, van der Meijden A. A non-lethal method for studying scorpion venom gland transcriptomes, with a review of potentially suitable taxa to which it can be applied. PLoS One 2021; 16:e0258712. [PMID: 34793470 PMCID: PMC8601437 DOI: 10.1371/journal.pone.0258712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Scorpion venoms are mixtures of proteins, peptides and small molecular compounds with high specificity for ion channels and are therefore considered to be promising candidates in the venoms-to-drugs pipeline. Transcriptomes are important tools for studying the composition and expression of scorpion venom. Unfortunately, studying the venom gland transcriptome traditionally requires sacrificing the animal and therefore is always a single snapshot in time. This paper describes a new way of generating a scorpion venom gland transcriptome without sacrificing the animal, thereby allowing the study of the transcriptome at various time points within a single individual. By comparing these venom-derived transcriptomes to the traditional whole-telson transcriptomes we show that the relative expression levels of the major toxin classes are similar. We further performed a multi-day extraction using our proposed method to show the possibility of doing a multiple time point transcriptome analysis. This allows for the study of patterns of toxin gene activation over time a single individual, and allows assessment of the effects of diet, season and other factors that are known or likely to influence intraindividual venom composition. We discuss the gland characteristics that may allow this method to be successful in scorpions and provide a review of other venomous taxa to which this method may potentially be successfully applied.
Collapse
Affiliation(s)
- Freek J. Vonk
- Naturalis Biodiversity Center, Leiden, The Netherlands
- Faculty of Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Mátyás A. Bittenbinder
- Naturalis Biodiversity Center, Leiden, The Netherlands
- Faculty of Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Harald M. I. Kerkkamp
- Naturalis Biodiversity Center, Leiden, The Netherlands
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | | | - John P. Archer
- CIBIO-InBIO, Biopolis, Universidade do Porto, Porto, Portugal
| | - Sandra Afonso
- CIBIO-InBIO, Biopolis, Universidade do Porto, Porto, Portugal
| | - Michael K. Richardson
- Animal Science and Health Cluster, Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Jeroen Kool
- Faculty of Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
30
|
dos Santos AT, Cruz GS, Baptista GR. Anti-inflammatory activities of arthropod peptides: a systematic review. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200152. [PMID: 34795699 PMCID: PMC8564866 DOI: 10.1590/1678-9199-jvatitd-2020-0152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/09/2021] [Indexed: 01/21/2023] Open
Abstract
Peptides obtained from different animal species have gained importance recently due to research that aims to develop biopharmaceuticals with therapeutic potential. In this sense, arthropod venoms have drawn attention, not only because of their toxicity but mainly for the search for molecules with various bioactivities, including anti-inflammatory activity. The purpose of the present study is to gather data available in the literature on new peptides derived from arthropod species with anti-inflammatory potential. This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. Studies on peptides from arthropods that display anti-inflammatory activity were retrieved from PubMed, Scopus, Web of Science, and Google Scholar databases. The bibliographic research started in 2020 and searched papers without a limit on the publication date. The articles were analyzed using a search string containing the following terms: "Peptides" and "Anti-inflammatory", in combinations such as "Ant", "Bee", "Wasp", "Crab", "Shrimp", "Scorpion", "Spider", "Tick" and "Centipedes". Besides, a search was carried out in the databases with the terms: "Peptides", "Antitumor", or "Anticancer", and "Arthropods". Articles that met the inclusion and exclusion criteria totalized 171, and these served for data extraction. Additionally, the present review included anti-inflammatory peptides with anticancer properties. Peptides with confirmed anti-inflammatory activity were from insects (ants, bees, and wasps), crustaceans (shrimp and crabs), arachnids (scorpions, spiders, and ticks), and centipedes. These arthropod peptides act mainly by decreasing pro-inflammatory cytokines as analyzed in vitro and in vivo. Some showed significant antineoplastic activity, working in essential cellular pathways against malignant neoplasms.
Collapse
Affiliation(s)
- Ariane Teixeira dos Santos
- Graduate Program in Pharmaceutical Sciences, School of Pharmacy, Dentistry and Nursing, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Gabriela Silva Cruz
- Graduate Program in Pharmaceutical Sciences, School of Pharmacy, Dentistry and Nursing, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Gandhi Rádis Baptista
- Graduate Program in Pharmaceutical Sciences, School of Pharmacy, Dentistry and Nursing, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| |
Collapse
|
31
|
A Pseudoscorpion's Promising Pinch: The venom of Chelifer cancroides contains a rich source of novel compounds. Toxicon 2021; 201:92-104. [PMID: 34416254 DOI: 10.1016/j.toxicon.2021.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
With pedipalps modified for venom injection, some pseudoscorpions possess a unique venom delivery system, which evolved independently from those of other arachnids like scorpions and spiders. Up to now, only a few studies have been focused on pseudoscorpion venom, which either identified a small fraction of venom compounds, or were based on solely transcriptomic approaches. Only one study addressed the bioactivity of pseudoscorpion venom. Here, we expand existing knowledge about pseudoscorpion venom by providing a comprehensive proteomic and transcriptomic analysis of the venom of Chelifer cancroides. We identified the first putative genuine toxins in the venom of C. cancroides and we showed that a large fraction of the venom comprises novel compounds. In addition, we tested the activity of the venom at specific ion channels for the first time. These tests demonstrate that the venom of C. cancroides causes inhibition of a voltage-gated insect potassium channel (Shaker IR) and modulates the inactivation process of voltage-gated sodium channels from Varroa destructor. For one of the smallest venomous animals ever studied, today's toolkits enabled a comprehensive venom analysis. This is demonstrated by allocating our identified venom compounds to more than half of the prominent ion signals in MALDI-TOF mass spectra of venom samples. The present study is a starting point for understanding the complex composition and activity of pseudoscorpion venom and provides a potential rich source of bioactive compounds useable for basic research and industrial application.
Collapse
|
32
|
Santos‐Filho NA, Righetto GM, Pereira MR, Piccoli JP, Almeida LMT, Leal TC, Camargo ILBC, Cilli EM. Effect of C‐terminal and N‐terminal dimerization and alanine scanning on antibacterial activity of the analogs of the peptide
p‐BthTX‐I. Pept Sci (Hoboken) 2021. [DOI: 10.1002/pep2.24243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
33
|
Sharma G, Braga CB, Chen KE, Jia X, Ramanujam V, Collins BM, Rittner R, Mobli M. Structural basis for the binding of the cancer targeting scorpion toxin, ClTx, to the vascular endothelia growth factor receptor neuropilin-1. Curr Res Struct Biol 2021; 3:179-186. [PMID: 34401749 PMCID: PMC8358460 DOI: 10.1016/j.crstbi.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/23/2021] [Accepted: 07/29/2021] [Indexed: 11/28/2022] Open
Abstract
Chlorotoxin (ClTx) is a 36-residue disulfide-rich peptide isolated from the venom of the scorpion Leiurus quinquestriatus. This peptide has been shown to selectively bind to brain tumours (gliomas), however, with conflicting reports regarding its direct cellular target. Recently, the vascular endothelial growth factor receptor, neuropilin-1 (NRP1) has emerged as a potential target of the peptide. Here, we sought to characterize the details of the binding of ClTx to the b1-domain of NRP1 (NRP1-b1) using solution state nuclear magnetic resonance (NMR) spectroscopy. The 3D structure of the isotope labelled peptide was solved using multidimensional heteronuclear NMR spectroscopy to produce a well-resolved structural ensemble. The structure points to three putative protein-protein interaction interfaces, two basic patches (R14/K15/K23 and R25/K27/R36) and a hydrophobic patch (F6/T7/T8/H10). The NRP1-b1 binding interface of ClTx was elucidated using 15N chemical shift mapping and included the R25/K27/R36 region of the peptide. The thermodynamics of binding was determined using isothermal titration calorimetry (ITC). In both NMR and ITC measurements, the binding was shown to be competitive with a known NRP1-b1 inhibitor. Finally, combining all of this data we generate a model of the ClTx:NRP1-b1 complex. The data shows that the peptide binds to the same region of NRP1 that is used by the SARS-CoV-2 virus for cell entry, however, via a non-canonical binding mode. Our results provide evidence for a non-standard NRP1 binding motif, while also providing a basis for further engineering of ClTx to generate peptides with improved NRP1 binding for future biomedical applications. Structural details of the binding of the scorpion toxin, chlorotoxin (ClTx) to the VEGF receptor neuropilin-1 (NRP1). ClTx was produced in its native fold and isotope labelled using a new, high-yield, bacterial expression system. Multidimensional heteronuclear NMR experiments reveal the high-resolution structure of ClTx and its binding to NRP1. ClTx binds to NRP1 via a non-canonical primary sequence that satisfies the receptor binding motif through its tertiary fold.
Collapse
Affiliation(s)
- Gagan Sharma
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | - Carolyne B. Braga
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
- Chemistry Institute, University of Campinas, P.O. Box 6154, 13083-970, Campinas, SP, Brazil
| | - Kai-En Chen
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Xinying Jia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | | | - Brett M. Collins
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Roberto Rittner
- Chemistry Institute, University of Campinas, P.O. Box 6154, 13083-970, Campinas, SP, Brazil
| | - Mehdi Mobli
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
- Corresponding author.
| |
Collapse
|
34
|
Drug Development Using Natural Toxins. Toxins (Basel) 2021; 13:toxins13060414. [PMID: 34207953 PMCID: PMC8230678 DOI: 10.3390/toxins13060414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
|
35
|
Lafnoune A, Lee SY, Heo JY, Gourja I, Darkaoui B, Abdelkafi-Koubaa Z, Chgoury F, Daoudi K, Chakir S, Cadi R, Mounaji K, Srairi-Abid N, Marrakchi N, Shum D, Seo HR, Oukkache N. Anti-Cancer Effect of Moroccan Cobra Naja haje Venom and Its Fractions against Hepatocellular Carcinoma in 3D Cell Culture. Toxins (Basel) 2021; 13:toxins13060402. [PMID: 34199838 PMCID: PMC8229680 DOI: 10.3390/toxins13060402] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/02/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer in adults, the fifth most common malignancy worldwide and the third leading cause of cancer related death. An alternative to the surgical treatments and drugs, such as sorafenib, commonly used in medicine is necessary to overcome this public health problem. In this study, we determine the anticancer effect on HCC of Moroccan cobra Naja haje venom and its fraction obtained by gel filtration chromatography against Huh7.5 cancer cell line. Cells were grown together with WI38 human fibroblast cells, LX2 human hepatic stellate cell line, and human endothelial cells (HUVEC) in MCTS (multi-cellular tumor spheroids) models. The hepatotoxicity of venom and its fractions were also evaluated using the normal hepatocytes cell line (Fa2N-4 cells). Our results showed that an anti HCC activity of Moroccan cobra Naja haje venom and, more specifically, the F7 fraction of gel filtration chromatography exhibited the greatest anti-hepatocellular carcinoma effect by decreasing the size of MCTS. This effect is associated with a low toxicity against normal hepatocytes. These results strongly suggest that the F7 fraction of Moroccan cobra Naja haje venom obtained by gel filtration chromatography possesses the ability to inhibit cancer cells proliferation. More research is needed to identify the specific molecule(s) responsible for the anticancer effect and investigate their mechanism of action.
Collapse
Affiliation(s)
- Ayoub Lafnoune
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
- Laboratoire Physiopathologie, Génétique Moléculaire & Biotechnologie, Faculté des Sciences Ain-Chock, Hassan II University of Casablanca, B.P 5366 Maarif, Casablanca 20000, Morocco; (R.C.); (K.M.)
| | - Su-Yeon Lee
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil Bundang-gu, Seong-nam-si 13488, Gyeonggi-do, Korea; (S.-Y.L.); (H.-R.S.)
| | - Jin-Yeong Heo
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil Bundang-gu, Seong-nam-si 13488, Gyeonggi-do, Korea; (J.-Y.H.); (D.S.)
| | - Imane Gourja
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
| | - Bouchra Darkaoui
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
- Laboratoire Physiopathologie, Génétique Moléculaire & Biotechnologie, Faculté des Sciences Ain-Chock, Hassan II University of Casablanca, B.P 5366 Maarif, Casablanca 20000, Morocco; (R.C.); (K.M.)
| | - Zaineb Abdelkafi-Koubaa
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, 13, Place Pasteur, Tunis 1002, Tunisia; (Z.A.-K.); (N.S.-A.); (N.M.)
| | - Fatima Chgoury
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
| | - Khadija Daoudi
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
- Laboratoire Physiopathologie, Génétique Moléculaire & Biotechnologie, Faculté des Sciences Ain-Chock, Hassan II University of Casablanca, B.P 5366 Maarif, Casablanca 20000, Morocco; (R.C.); (K.M.)
| | - Salma Chakir
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
| | - Rachida Cadi
- Laboratoire Physiopathologie, Génétique Moléculaire & Biotechnologie, Faculté des Sciences Ain-Chock, Hassan II University of Casablanca, B.P 5366 Maarif, Casablanca 20000, Morocco; (R.C.); (K.M.)
| | - Khadija Mounaji
- Laboratoire Physiopathologie, Génétique Moléculaire & Biotechnologie, Faculté des Sciences Ain-Chock, Hassan II University of Casablanca, B.P 5366 Maarif, Casablanca 20000, Morocco; (R.C.); (K.M.)
| | - Najet Srairi-Abid
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, 13, Place Pasteur, Tunis 1002, Tunisia; (Z.A.-K.); (N.S.-A.); (N.M.)
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, 13, Place Pasteur, Tunis 1002, Tunisia; (Z.A.-K.); (N.S.-A.); (N.M.)
| | - David Shum
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil Bundang-gu, Seong-nam-si 13488, Gyeonggi-do, Korea; (J.-Y.H.); (D.S.)
| | - Haeng-Ran Seo
- Cancer Biology Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil Bundang-gu, Seong-nam-si 13488, Gyeonggi-do, Korea; (S.-Y.L.); (H.-R.S.)
| | - Naoual Oukkache
- Laboratoire des Venins et Toxines, Département de Recherche, Institut Pasteur du Maroc, 1, Place Louis Pasteur, Casablanca 20360, Morocco; (A.L.); (I.G.); (B.D.); (F.C.); (K.D.); (S.C.)
- Correspondence:
| |
Collapse
|
36
|
Tang X, Yu D, Wang H, Meng W, Lei Z, Zhai Y, Wang Y, Wang X. Biochemical and cytotoxic evaluation of latroeggtoxin-VI against PC12 cells. J Biochem Mol Toxicol 2021; 35:e22825. [PMID: 34047418 DOI: 10.1002/jbt.22825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/27/2021] [Accepted: 05/18/2021] [Indexed: 01/03/2023]
Abstract
Latroeggtoxin-VI (LETX-VI) is a peptide neurotoxin discovered from Latrodectus tredecimguttatus eggs. In the current study, the action features of the neurotoxin on PC12 cells were systematically investigated. LETX-VI could promote dopamine release from PC12 cells in the absence and presence of Ca2+, involving an even more complex action mechanism in the presence of Ca2+ and when the treatment time was longer. Although LETX-VI enchanced the autophagy and secretion activity in PC 12 cells, it showed no remarkable influence on the proliferation, cell cycle, apoptosis and ultrastructure of the cells. Pulldown combined with CapLC-MS/MS analysis suggested that LETX-VI affected PC12 cells by interacting with multiple proteins involved in the metabolism, transport, and release of neurotransmitters, particularly dopamine. The low cytotoxicity and effective regulatory action of LETX-VI on PC12 cells suggest the potential of the active peptide in the development of drugs for the treatment of some dopamine-related psychotic diseases and cancers.
Collapse
Affiliation(s)
- Xiaochao Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Dianmei Yu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Haiyan Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Wenwen Meng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Zhixiang Lei
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Yiwen Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Ying Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xianchun Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Protein Chemistry Laboratory, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
37
|
Bagheri-Ziari S, Shahbazzadeh D, Sardari S, Sabatier JM, Pooshang Bagheri K. Discovery of a New Analgesic Peptide, Leptucin, from the Iranian Scorpion, Hemiscorpius lepturus. Molecules 2021; 26:molecules26092580. [PMID: 33925223 PMCID: PMC8124257 DOI: 10.3390/molecules26092580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023] Open
Abstract
Hemiscorpius lepturus scorpion stings do not induce considerable pain based on epidemiological surveys conducted in the southwest part of Iran. Accordingly, this study was aimed to identify the analgesic molecule in H. lepturus venom by analyzing a cDNA library of the scorpion venom gland looking for sequences having homology with known animal venom analgesic peptides. The analgesic molecule is a cysteine rich peptide of 55 amino acids. the synthetic peptide was deprotected and refolded. RP-HPLC, Ellman's, and DLS assays confirmed the refolding accuracy. Circular dichroism (CD) showed helix and beta sheet contents. This peptide, called leptucin, demonstrated 95% analgesic activity at the dose of 0.48 mg/kg in hot plate assay. Leptucin at the doses of 0.32, 0.48, and 0.64 mg/kg showed 100% activity in thermal tail flick test. No hemolysis or cytotoxicity was observed at 8 and 16 µg. Histopathology evaluations indicated no hepatotoxicity, nephrotoxicity, and cardiotoxicity. We thus report that leptucin is the analgesic agent of H. lepturus venom. Regarding the high in vivo efficacy of leptucin and the fact it shows no observable toxicity, it could be suggested as a drug lead in a preclinical study of acute pain as well as the study of its mechanism of action.
Collapse
Affiliation(s)
- Sedigheh Bagheri-Ziari
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.B.-Z.); (D.S.)
| | - Delavar Shahbazzadeh
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.B.-Z.); (D.S.)
| | - Soroush Sardari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Jean-Marc Sabatier
- Institute of NeuroPhysiopathology (INP), Faculté de Pharmacie, Université d’Aix-Marseille, UMR 7051, 27 Bd Jean Moulin, CEDEX, 13385 Marseille, France;
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.B.-Z.); (D.S.)
- Correspondence:
| |
Collapse
|
38
|
Samianifard M, Nazari A, Tahoori F, Mohamadpour Dounighi N. Proteome Analysis of Toxic Fractions of Iranian Cobra (Naja naja Oxiana) Snake Venom Using Two-Dimensional Electrophoresis and Mass Spectrometry. ARCHIVES OF RAZI INSTITUTE 2021; 76:127-138. [PMID: 33818965 DOI: 10.22092/ari.2020.128766.1428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/14/2020] [Indexed: 09/30/2022]
Abstract
Snake venoms are mostly composed of various proteins and peptides with toxicity and pharmacological effects depending on their geographical sources. Naja naja oxiana is one of the most medically important venomous snakes in Iran and Central Asia. The bite of this type of snake can cause severe pain and swelling, as well as neurotoxicity. Without medical treatment, symptoms quickly worsen and death can occur soon. A detailed understanding of venom components can provide new insight into the production of antivenom against toxic agents instead of crude venom. Specific antibodies against toxic fractions are of utmost importance in neutralizing crude venom. Therefore, the proteome profile of these fractions of Naja naja oxidana venom was analyzed using fractionation by gel filtration, two-dimensional electrophoresis, mass spectrometry, and data mining. Base on the results, in total, 32 spots were detected and categorized into three protein families, namely three-finger toxin (3FTx), phospholipase, and Cysteine-rich secretory proteins (CRISP). These proteins consist of more than 70% crude venom all with a molecular weight below 25 kDa. The 3FTx as a highly diverse constituent in the venom of Naja species was in large quantity in this district. Short-chain neurotoxins, including short neurotoxin, cytotoxin, and muscarinic toxin-like protein, were in abundance, respectively. In conclusion, the recognition of toxic fractions of Naja naja oxiana in this region could be of great help in the production of an effective antivenom against similar compositions. It can also help the medical care department to find out the clinical sign of cobra venom. To the best of our knowledge, this was the first study to report the proteomic of toxic fractions of Naja naja oxiana in Iran.
Collapse
Affiliation(s)
- M Samianifard
- Department of Proteomics-Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - A Nazari
- Department of Proteomics-Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.,Department of Proteomics-Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - F Tahoori
- Department of Human Bacterial Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - N Mohamadpour Dounighi
- Department of Venomous Animal, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
39
|
Patra A, Mukherjee AK. Assessment of snakebite burdens, clinical features of envenomation, and strategies to improve snakebite management in Vietnam. Acta Trop 2021; 216:105833. [PMID: 33485869 DOI: 10.1016/j.actatropica.2021.105833] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/16/2022]
Abstract
The sheer paucity of scientific documentation of herpetofauna in Vietnam and the rudimentary healthcare response to snakebite have stimulated this review. Over six decades of data culled from public data bases and search engines, have been used to assess snakebite burdens, clinical features of envenomation, and strategies for snakebite management in Vietnam. In addition, biochemical and proteomic analyses to decipher venom composition, rapid analytical techniques to be used for clinical diagnosis of snakebite in Vietnam have been discussed in detail. The assessment of efficacy, safety, and quality of commercial antivenom produced in Vietnam and improvement of antivenom production to meet the national requirement has been critically examined. It is apparent that snake bite incidence in Vietnam is exacerbated by mismatch in demand and supply of antivenom therapy, insufficient medical facilities, preference for traditional healers and poor management of clinical records. The impediments arising from geographical and species-specific variation in venom composition can be overcome by the 'Omics approach', and scientific documentation of pathophysiological manifestations post envenomation. The development of next generation of therapeutics, encouraging clinical research, novel approaches and social awareness against snakebite and its treatments have been suggested to significantly reduce the snakebite mortality and morbidity in this region.
Collapse
Affiliation(s)
- Aparup Patra
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India.
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; Institute of Advanced Study in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India.
| |
Collapse
|
40
|
Coulter-Parkhill A, McClean S, Gault VA, Irwin N. Therapeutic Potential of Peptides Derived from Animal Venoms: Current Views and Emerging Drugs for Diabetes. Clin Med Insights Endocrinol Diabetes 2021; 14:11795514211006071. [PMID: 34621137 PMCID: PMC8491154 DOI: 10.1177/11795514211006071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic potential of venom-derived drugs is evident today. Currently, several significant drugs are FDA approved for human use that descend directly from animal venom products, with others having undergone, or progressing through, clinical trials. In addition, there is growing awareness of the important cosmeceutical application of venom-derived products. The success of venom-derived compounds is linked to their increased bioactivity, specificity and stability when compared to synthetically engineered compounds. This review highlights advancements in venom-derived compounds for the treatment of diabetes and related disorders. Exendin-4, originating from the saliva of Gila monster lizard, represents proof-of-concept for this drug discovery pathway in diabetes. More recent evidence emphasises the potential of venom-derived compounds from bees, cone snails, sea anemones, scorpions, snakes and spiders to effectively manage glycaemic control. Such compounds could represent exciting exploitable scaffolds for future drug discovery in diabetes, as well as providing tools to allow for a better understanding of cell signalling pathways linked to insulin secretion and metabolism.
Collapse
Affiliation(s)
| | | | - Victor A Gault
- Diabetes Research Group, Ulster University, Coleraine, UK
| | - Nigel Irwin
- Diabetes Research Group, Ulster University, Coleraine, UK
| |
Collapse
|
41
|
Adamude FA, Dingwoke EJ, Abubakar MS, Ibrahim S, Mohamed G, Klein A, Sallau AB. Proteomic analysis of three medically important Nigerian Naja (Naja haje, Naja katiensis and Naja nigricollis) snake venoms. Toxicon 2021; 197:24-32. [PMID: 33775665 DOI: 10.1016/j.toxicon.2021.03.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022]
Abstract
Proteomics technologies enable a comprehensive study of complex proteins and their functions. The venom proteomes of three medically important Nigerian Elapidae snakes Naja haje, Naja katiensis and Naja nigricollis was studied using HILIC coupled with LC-MS/MS analysis. Results revealed a total of 57, 55, and 46 proteins in the venoms of N. haje, N. katiensis, and N. nigricollis, respectively, with molecular mass ranging between 5 and 185 kDa. These snakes have 38 common proteins in addition to 3 uncommon proteins: actiflagelin, cathelicidin, and cystatin identified in their venoms. The identified proteins belonged to 14 protein families in N. haje and N. katiensis, and 12 protein families in N. nigricollis. Of the total venom proteins, 3FTx was the most abundant protein family, constituting 52% in N. haje and N. katiensis, and 41% in N. nigricollis, followed by PLA2, constituting 37% in N. nigricollis, 26% in N. haje, and 24% in N. katiensis. Other protein families, including LAAO, CRISPs, VEGF, PLB, CVF, SVMP, SVH, AMP, PI, Globin, Actin, and C-type lectins, were also detected, although, at very low abundances. Quantification of the relative abundance of each protein revealed that alpha and beta fibrinogenase and PLA2, which constituted 18-26% of the total proteome, were the most abundant. The 3 uncommon proteins have no known function in snake venom. However, actiflagelin activates sperm motility; cystatin inhibits angiogenesis, while cathelicidin exerts antimicrobial effects. The three Nigerian Naja genus proteomes displayed 70% similarity in composition, which suggests the possibility of formulating antivenom that may cross-neutralise the venoms of cobra species found in Nigeria. These data provide insights into clinically relevant peptides/proteins present in the venoms of these snakes. Data are available via ProteomeXchange with identifier PXD024627.
Collapse
Affiliation(s)
- Fatima Amin Adamude
- Department of Biochemistry, Faculty of Sciences, Federal University of Lafia, Nasarawa State, Nigeria; Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Emeka John Dingwoke
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria; Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
| | - Mujitaba Suleiman Abubakar
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria; Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Sani Ibrahim
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria; Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Gadija Mohamed
- Agri-Food Systems and Omics, Post-Harvest and Agro-Processing Technologies, Agricultural Research Council, Infrutec-Nietvoorbij, Stellenbosch, 7599, South Africa
| | - Ashwil Klein
- Proteomics Research Unit, Department of Biotechnology, Faculty of Natural Sciences, University of Western Cape, South Africa
| | - Abdullahi Balarabe Sallau
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria; Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| |
Collapse
|
42
|
Abd El-Aziz TM, Soares AG, Stockand JD. Advances in venomics: Modern separation techniques and mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1160:122352. [PMID: 32971366 PMCID: PMC8174749 DOI: 10.1016/j.jchromb.2020.122352] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/31/2022]
Abstract
Snake venoms are complex chemical mixtures of biologically active proteins and non-protein components. Toxins have a wide range of targets and effects to include ion channels and membrane receptors, and platelet aggregation and platelet plug formation. Toxins target these effectors and effects at high affinity and selectivity. From a pharmacological perspective, snake venom compounds are a valuable resource for drug discovery and development. However, a major challenge to drug discovery using snake venoms is isolating and analyzing the bioactive proteins and peptides in these complex mixtures. Getting molecular information from complex mixtures such as snake venoms requires proteomic analyses, generally combined with transcriptomic analyses of venom glands. The present review summarizes current knowledge and highlights important recent advances in venomics with special emphasis on contemporary separation techniques and bioinformatics that have begun to elaborate the complexity of snake venoms. Several analytical techniques such as two-dimensional gel electrophoresis, RP-HPLC, size exclusion chromatography, ion exchange chromatography, MALDI-TOF-MS, and LC-ESI-QTOF-MS have been employed in this regard. The improvement of separation approaches such as multidimensional-HPLC, 2D-electrophoresis coupled to soft-ionization (MALDI and ESI) mass spectrometry has been critical to obtain an accurate picture of the startling complexity of venoms. In the case of bioinformatics, a variety of software tools such as PEAKS also has been used successfully. Such information gleaned from venomics is important to both predicting and resolving the biological activity of the active components of venoms, which in turn is key for the development of new drugs based on these venom components.
Collapse
Affiliation(s)
- Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900, USA; Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt.
| | - Antonio G Soares
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900, USA
| | - James D Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900, USA
| |
Collapse
|
43
|
Almeida AACD, Ferreira JRDO, de Carvalho RBF, Rizzo MDS, Lopes LDS, Dittz D, Castro E Souza JMD, Ferreira PMP. Non-clinical toxicity of (+)-limonene epoxide and its physio-pharmacological properties on neurological disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2301-2314. [PMID: 32653979 DOI: 10.1007/s00210-020-01943-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022]
Abstract
The compound (+)-limonene epoxide has antioxidant, anxiolytic, and antihelminthic properties. However, investigations to determine its long-term exposure were not performed. We investigated the systemic toxicological profile after chronic exposure as well as the antidepressant and antiepileptic potentialities of (+)-limonene epoxide on mice. Initially, we evaluated acute toxicity on Artemia salina nauplii and cytotoxicity on mice erythrocytes and peripheral blood mononuclear cells (PBMC). Aftterwards, mice were chronically treated for 120 days by gavage with (+)-limonene epoxide (25, 50, and 75 mg/kg/day) and this exposure was assessed by pathophysiological measurements. For antidepressant and anticonvulsivant analysis, we performed the forced swimming and tail suspension protocols and pentylenetetrazol- and picrotoxin-induced seizures, respectively. (+)-Limonene epoxide showed a LC50 value of 318.7 μg/mL on A. salina shrimps, caused lysis of red blood cells at higher concentrations only but did not show cytotoxicity on PMBC, which suggests pharmacological safety if plasma concentrations do not exceed 100 μg/mL. Macroscopic, hematological, clinical chemistry, and nutritional changes were not detected, though focal areas of hepatic necrosis, inflammatory infiltrate, and karyolysis have been detected at 75 mg/kg/day. The compound inhibited the developing of pentylenetetrazol- and picrotoxin-induced seizures, decreased deaths, and reduced immobility times, mainly at 75 mg/kg. So, it reversed reserpine effects, suggesting antidepressant effects should be linked to serotonergic and/or adrenergic transmission. It is feasible that (+)-limonene epoxide plays a benzodiazepine-like anticonvulsive action and may be also recommended as an antidote for poisonings caused by central depressants.
Collapse
Affiliation(s)
- Antonia Amanda Cardoso de Almeida
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, 64049-550, Brazil
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, Universitária Avenue, Ininga, Teresina, Piauí, 64049-550, Brazil
| | | | | | | | - Luciano da Silva Lopes
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, 64049-550, Brazil
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, Universitária Avenue, Ininga, Teresina, Piauí, 64049-550, Brazil
| | - Dalton Dittz
- Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - João Marcelo de Castro E Souza
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, 64049-550, Brazil
- Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, 64049-550, Brazil.
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, Universitária Avenue, Ininga, Teresina, Piauí, 64049-550, Brazil.
| |
Collapse
|
44
|
Gopcevic K, Karadzic I, Izrael-Zivkovic L, Medic A, Isakovic A, Popović M, Kekic D, Stanojkovic T, Hozic A, Cindric M. Study of the venom proteome of Vipera ammodytes ammodytes (Linnaeus, 1758): A qualitative overview, biochemical and biological profiling. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 37:100776. [PMID: 33197857 DOI: 10.1016/j.cbd.2020.100776] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/13/2020] [Accepted: 11/04/2020] [Indexed: 12/23/2022]
Abstract
Vipera ammodytes (Va), is the European venomous snake of the greatest medical importance. We analyzed whole venom proteome of the subspecies V. ammodytes ammodytes (Vaa) from Serbia for the first time using the shotgun proteomics approach and identified 99 proteins belonging to four enzymatic families: serine protease (SVSPs), L-amino acid oxidase (LAAOs), metalloproteinases (SVMPs), group II phospholipase (PLA2s), and five nonenzymatic families: cysteine-rich secretory proteins (CRISPs), C-type lectins (snaclecs), growth factors -nerve (NGFs) and vascular endothelium (VEGFs), and Kunitz-type protease inhibitors (SPIs). Considerable enzymatic activity of LAAO, SVSPs, and SVMPs and a high acidic PLA2 activity was measured implying potential of Vaa to produce haemotoxic, myotoxic, neuro and cardiotoxic effects. Moreover, significant antimicrobial activity of Vaa venom against Gram-negative (Klebsiella pneumoniae, Pseudomonas aeruginosa) and Gram-positive bacteria (Staphylococcus aureus) was found. The crude venom shows considerable potential cytotoxic activity on the C6 and HL60 and a moderate level of potency on B16 cell lines. HeLa cells showed the same sensitivity, while DU 145 and PC-3 are less sensitive than as normal cell line. Our data demonstrated a high complexity of Vaa and considerable enzymatic, antibacterial and cytotoxic activity, implying a great medical potential of Vaa venom as a promising source for new antibacterial and cytostatic agents.
Collapse
Affiliation(s)
- Kristina Gopcevic
- Department of Chemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia.
| | - Ivanka Karadzic
- Department of Chemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Lidija Izrael-Zivkovic
- Department of Chemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Ana Medic
- Department of Chemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Isakovic
- Department of Medical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marjan Popović
- Department of Medical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dusan Kekic
- Department of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Amela Hozic
- Ruđer Bošković Institute, Proteomics and Mass Spectrometry, Zagreb, Croatia
| | - Mario Cindric
- Ruđer Bošković Institute, Proteomics and Mass Spectrometry, Zagreb, Croatia
| |
Collapse
|
45
|
Mendel HC, Kaas Q, Muttenthaler M. Neuropeptide signalling systems - An underexplored target for venom drug discovery. Biochem Pharmacol 2020; 181:114129. [PMID: 32619425 PMCID: PMC7116218 DOI: 10.1016/j.bcp.2020.114129] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 01/06/2023]
Abstract
Neuropeptides are signalling molecules mainly secreted from neurons that act as neurotransmitters or peptide hormones to affect physiological processes and modulate behaviours. In humans, neuropeptides are implicated in numerous diseases and understanding their role in physiological processes and pathologies is important for therapeutic development. Teasing apart the (patho)physiology of neuropeptides remains difficult due to ligand and receptor promiscuity and the complexity of the signalling pathways. The current approach relies on a pharmacological toolbox of agonists and antagonists displaying high selectivity for independent receptor subtypes, with the caveat that only few selective ligands have been discovered or developed. Animal venoms represent an underexplored source for novel receptor subtype-selective ligands that could aid in dissecting human neuropeptide signalling systems. Multiple endogenous-like neuropeptides as well as peptides acting on neuropeptide receptors are present in venoms. In this review, we summarise current knowledge on neuropeptides and discuss venoms as a source for ligands targeting neuropeptide signalling systems.
Collapse
Affiliation(s)
- Helen C Mendel
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Quentin Kaas
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Markus Muttenthaler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia; University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Vienna, Austria.
| |
Collapse
|
46
|
Herzig V, Cristofori-Armstrong B, Israel MR, Nixon SA, Vetter I, King GF. Animal toxins - Nature's evolutionary-refined toolkit for basic research and drug discovery. Biochem Pharmacol 2020; 181:114096. [PMID: 32535105 PMCID: PMC7290223 DOI: 10.1016/j.bcp.2020.114096] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/27/2022]
Abstract
Venomous animals have evolved toxins that interfere with specific components of their victim's core physiological systems, thereby causing biological dysfunction that aids in prey capture, defense against predators, or other roles such as intraspecific competition. Many animal lineages evolved venom systems independently, highlighting the success of this strategy. Over the course of evolution, toxins with exceptional specificity and high potency for their intended molecular targets have prevailed, making venoms an invaluable and almost inexhaustible source of bioactive molecules, some of which have found use as pharmacological tools, human therapeutics, and bioinsecticides. Current biomedically-focused research on venoms is directed towards their use in delineating the physiological role of toxin molecular targets such as ion channels and receptors, studying or treating human diseases, targeting vectors of human diseases, and treating microbial and parasitic infections. We provide examples of each of these areas of venom research, highlighting the potential that venom molecules hold for basic research and drug development.
Collapse
Affiliation(s)
- Volker Herzig
- School of Science & Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| | | | - Mathilde R Israel
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Samantha A Nixon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
47
|
Ryan RYM, Lutzky VP, Herzig V, Smallwood TB, Potriquet J, Wong Y, Masci P, Lavin MF, King GF, Lopez JA, Ikonomopoulou MP, Miles JJ. Venom of the Red-Bellied Black Snake Pseudechis porphyriacus Shows Immunosuppressive Potential. Toxins (Basel) 2020; 12:toxins12110674. [PMID: 33114591 PMCID: PMC7693913 DOI: 10.3390/toxins12110674] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023] Open
Abstract
Venoms act with remarkable specificity upon a broad diversity of physiological targets. Venoms are composed of proteins, peptides, and small molecules, providing the foundation for the development of novel therapeutics. This study assessed the effect of venom from the red-bellied black snake (Pseudechis porphyriacus) on human primary leukocytes using bead-based flow cytometry, mixed lymphocyte reaction, and cell viability assays. We show that venom treatment had a significant immunosuppressive effect, inhibiting the secretion of interleukin (IL)-2 and tumor necrosis factor (TNF) from purified human T cells by 90% or greater following stimulation with mitogen (phorbol 12-myristate 13-acetate and ionomycin) or via cluster of differentiation (CD) receptors, CD3/CD28. In contrast, venom treatment did not inhibit TNF or IL-6 release from antigen-presenting cells stimulated with lipopolysaccharide. The reduced cytokine release from T cells was not associated with inhibition of T cell proliferation or reduction of cell viability, consistent with an anti-inflammatory mechanism unrelated to the cell cycle. Deconvolution of the venom using reverse-phase HPLC identified four fractions responsible for the observed immunosuppressive activity. These data suggest that compounds from P. porphyriacus venom may be potential drug leads for T cell-associated conditions such as graft versus host disease, rheumatoid arthritis, and inflammatory bowel disease.
Collapse
Affiliation(s)
- Rachael Y. M. Ryan
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4878, Australia;
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD 4870, Australia
- School of Environment and Sciences, Griffith University, Nathan, QLD 4111, Australia;
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (V.P.L.); (T.B.S.); (M.P.I.)
- Correspondence: (R.Y.M.R.); (J.J.M.)
| | - Viviana P. Lutzky
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (V.P.L.); (T.B.S.); (M.P.I.)
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; (V.H.); (G.F.K.)
- GeneCology Research Centre, School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
| | - Taylor B. Smallwood
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (V.P.L.); (T.B.S.); (M.P.I.)
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | | | - Yide Wong
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4878, Australia;
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD 4870, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD 4878, Australia
| | - Paul Masci
- Translational Research Institute, Brisbane, QLD 4102, Australia;
| | - Martin F. Lavin
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia;
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; (V.H.); (G.F.K.)
| | - J. Alejandro Lopez
- School of Environment and Sciences, Griffith University, Nathan, QLD 4111, Australia;
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (V.P.L.); (T.B.S.); (M.P.I.)
| | - Maria P. Ikonomopoulou
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (V.P.L.); (T.B.S.); (M.P.I.)
- Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, 28049 Madrid, Spain
| | - John J. Miles
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4878, Australia;
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD 4870, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD 4878, Australia
- Correspondence: (R.Y.M.R.); (J.J.M.)
| |
Collapse
|
48
|
Functional Mining of the Crotalus Spp. Venom Protease Repertoire Reveals Potential for Chronic Wound Therapeutics. Molecules 2020; 25:molecules25153401. [PMID: 32731325 PMCID: PMC7435869 DOI: 10.3390/molecules25153401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/03/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic wounds are a major health problem that cause millions of dollars in expenses every year. Among all the treatments used, active wound treatments such as enzymatic treatments represent a cheaper and specific option with a fast growth category in the market. In particular, bacterial and plant proteases have been employed due to their homology to human proteases, which drive the normal wound healing process. However, the use of these proteases has demonstrated results with low reproducibility. Therefore, alternative sources of proteases such as snake venom have been proposed. Here, we performed a functional mining of proteases from rattlesnakes (Crotalus ornatus, C. molossus nigrescens, C. scutulatus, and C. atrox) due to their high protease predominance and similarity to native proteases. To characterize Crotalus spp. Proteases, we performed different protease assays to measure and confirm the presence of metalloproteases and serine proteases, such as the universal protease assay and zymography, using several substrates such as gelatin, casein, hemoglobin, L-TAME, fibrinogen, and fibrin. We found that all our venom extracts degraded casein, gelatin, L-TAME, fibrinogen, and fibrin, but not hemoglobin. Crotalus ornatus and C. m. nigrescens extracts were the most proteolytic venoms among the samples. Particularly, C. ornatus predominantly possessed low molecular weight proteases (P-I metalloproteases). Our results demonstrated the presence of metalloproteases capable of degrading gelatin (a collagen derivative) and fibrin clots, whereas serine proteases were capable of degrading fibrinogen-generating fibrin clots, mimicking thrombin activity. Moreover, we demonstrated that Crotalus spp. are a valuable source of proteases that can aid chronic wound-healing treatments.
Collapse
|
49
|
Liu ZC, Liang JY, Lan XQ, Li T, Zhang JR, Zhao F, Li G, Chen PY, Zhang Y, Lee WH, Zhao F. Comparative analysis of diverse toxins from a new pharmaceutical centipede, Scolopendra mojiangica. Zool Res 2020; 41:138-147. [PMID: 31945809 PMCID: PMC7109010 DOI: 10.24272/j.issn.2095-8137.2020.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
As the oldest venomous animals, centipedes use their venom as a weapon to attack prey and for protection. Centipede venom, which contains many bioactive and pharmacologically active compounds, has been used for centuries in Chinese medicine, as shown by ancient records. Based on comparative analysis, we revealed the diversity of and differences in centipede toxin-like molecules between Scolopendra mojiangica, a substitute pharmaceutical material used in China, and S. subspinipes mutilans. More than 6 000 peptides isolated from the venom were identified by electrospray ionization-tandem mass spectrometry (ESI-MS/MS) and inferred from the transcriptome. As a result, in the proteome of S. mojiangica, 246 unique proteins were identified: one in five were toxin-like proteins or putative toxins with unknown function, accounting for a lower percentage of total proteins than that in S. mutilans. Transcriptome mining identified approximately 10 times more toxin-like proteins, which can characterize the precursor structures of mature toxin-like peptides. However, the constitution and quantity of the toxin transcripts in these two centipedes were similar. In toxicity assays, the crude venom showed strong insecticidal and hemolytic activity. These findings highlight the extensive diversity of toxin-like proteins in S. mojiangica and provide a new foundation for the medical-pharmaceutical use of centipede toxin-like proteins.
Collapse
Affiliation(s)
- Zi-Chao Liu
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Engineering Research Center for Exploitation and Utilization of Leech Resources in Universities of Yunnan Province, School of Agronomy and Life Sciences, Kunming University, Kunming, Yunnan 650214, China
| | - Jin-Yang Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xin-Qiang Lan
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Tao Li
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Key Laboratory of Active Molecules and Drug Development, Puer University, Puer, Yunnan 665000, China
| | - Jia-Rui Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Nanshan College, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Fang Zhao
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Key Laboratory of Active Molecules and Drug Development, Puer University, Puer, Yunnan 665000, China.,Institute of Comparative Study of Traditional Materia Medica, Institute of Integrative Medicine of Fudan University, Shanghai 200032, China
| | - Geng Li
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Key Laboratory of Active Molecules and Drug Development, Puer University, Puer, Yunnan 665000, China
| | - Pei-Yi Chen
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Key Laboratory of Active Molecules and Drug Development, Puer University, Puer, Yunnan 665000, China
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| | - Wen-Hui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail: leewh@mail. kiz.ac.cn
| | - Feng Zhao
- Key Laboratory of Ethnic Medical Resources Research and Southeast Asian International Cooperation of Yunnan Universities, Department of Biology and Chemistry, Puer University, Puer, Yunnan 665000, China.,Key Laboratory of Active Molecules and Drug Development, Puer University, Puer, Yunnan 665000, China.,Institute of Comparative Study of Traditional Materia Medica, Institute of Integrative Medicine of Fudan University, Shanghai 200032, China. E-mail:
| |
Collapse
|
50
|
Bordon KDCF, Cologna CT, Fornari-Baldo EC, Pinheiro-Júnior EL, Cerni FA, Amorim FG, Anjolette FAP, Cordeiro FA, Wiezel GA, Cardoso IA, Ferreira IG, de Oliveira IS, Boldrini-França J, Pucca MB, Baldo MA, Arantes EC. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front Pharmacol 2020; 11:1132. [PMID: 32848750 PMCID: PMC7396678 DOI: 10.3389/fphar.2020.01132] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Animal poisons and venoms are comprised of different classes of molecules displaying wide-ranging pharmacological activities. This review aims to provide an in-depth view of toxin-based compounds from terrestrial and marine organisms used as diagnostic tools, experimental molecules to validate postulated therapeutic targets, drug libraries, prototypes for the design of drugs, cosmeceuticals, and therapeutic agents. However, making these molecules applicable requires extensive preclinical trials, with some applications also demanding clinical trials, in order to validate their molecular target, mechanism of action, effective dose, potential adverse effects, as well as other fundamental parameters. Here we go through the pitfalls for a toxin-based potential therapeutic drug to become eligible for clinical trials and marketing. The manuscript also presents an overview of the current picture for several molecules from different animal venoms and poisons (such as those from amphibians, cone snails, hymenopterans, scorpions, sea anemones, snakes, spiders, tetraodontiformes, bats, and shrews) that have been used in clinical trials. Advances and perspectives on the therapeutic potential of molecules from other underexploited animals, such as caterpillars and ticks, are also reported. The challenges faced during the lengthy and costly preclinical and clinical studies and how to overcome these hindrances are also discussed for that drug candidates going to the bedside. It covers most of the drugs developed using toxins, the molecules that have failed and those that are currently in clinical trials. The article presents a detailed overview of toxins that have been used as therapeutic agents, including their discovery, formulation, dosage, indications, main adverse effects, and pregnancy and breastfeeding prescription warnings. Toxins in diagnosis, as well as cosmeceuticals and atypical therapies (bee venom and leech therapies) are also reported. The level of cumulative and detailed information provided in this review may help pharmacists, physicians, biotechnologists, pharmacologists, and scientists interested in toxinology, drug discovery, and development of toxin-based products.
Collapse
Affiliation(s)
- Karla de Castro Figueiredo Bordon
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Takeno Cologna
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ernesto Lopes Pinheiro-Júnior
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Gobbi Amorim
- Postgraduate Program in Pharmaceutical Sciences, Vila Velha University, Vila Velha, Brazil
| | | | - Francielle Almeida Cordeiro
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gisele Adriano Wiezel
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Iara Aimê Cardoso
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isabela Gobbo Ferreira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora Sousa de Oliveira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Mateus Amaral Baldo
- Health and Science Institute, Paulista University, São José do Rio Pardo, Brazil
| | - Eliane Candiani Arantes
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|